Next Article in Journal
Applications of Exosomes in Diagnosing Muscle Invasive Bladder Cancer
Next Article in Special Issue
Development of Biocompatible Ciprofloxacin–Gold Nanoparticle Coated Sutures for Surgical Site Infections
Previous Article in Journal
Manufacturability of a Tetraethyl Orthosilicate-Based Hydrogel for Use as a Single Application Otitis Externa Therapeutic
Previous Article in Special Issue
Wound-Healing Promotion and Anti-Inflammatory Properties of Carvacrol Prodrugs/Hyaluronic Acid Formulations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hybrid Azine Derivatives: A Useful Approach for Antimicrobial Therapy

by
Dorina Amariucai-Mantu
1,
Violeta Mangalagiu
2,*,
Iustinian Bejan
1,2,
Aculina Aricu
1,3 and
Ionel I. Mangalagiu
1,2,*
1
Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 11 Carol 1st Bvd, 700506 Iasi, Romania
2
CERNESIM Centre, Institute of Interdisciplinary Research, Alexandru Ioan Cuza University of Iasi, 11 Carol I, 700506 Iasi, Romania
3
Chemistry of Natural and Biologically Active Compounds Laboratory, Institute of Chemistry, 3 Academiei Str., MD-2028 Chisinau, Moldova
*
Authors to whom correspondence should be addressed.
Pharmaceutics 2022, 14(10), 2026; https://doi.org/10.3390/pharmaceutics14102026
Submission received: 22 August 2022 / Revised: 14 September 2022 / Accepted: 16 September 2022 / Published: 23 September 2022

Abstract

:
Nowadays, infectious diseases caused by microorganisms are a major threat to human health, mostly because of drug resistance, multi-drug resistance and extensive-drug-resistance phenomena to microbial pathogens. During the last few years, obtaining hybrid azaheterocyclic drugs represents a powerful and attractive approach in modern antimicrobial therapy with very promising results including overcoming microbial drug resistance. The emphasis of this review is to notify the scientific community about the latest recent advances from the last five years in the field of hybrid azine derivatives with antimicrobial activity. The review is divided according to the main series of six-member ring azaheterocycles with one nitrogen atom and their fused analogs. In each case, the main essential data concerning synthesis and antimicrobial activity are presented.

Graphical Abstract

1. Introduction

According to the WHO, infectious diseases caused by microorganisms represent a major threat that affects society and human health, exerting great pressure on health systems, individuals and communities [1]. In particular, overconsumption and widespread use and misuse of antimicrobial agents have resulted in the emergence of drug resistance, multi-drug resistance and extensive-drug-resistance phenomena to microbial pathogens and many other drawbacks (toxicity and non specificity of drugs, high prices, etc.). So far, searching for new chemical entities with improved antimicrobial properties remains a very challenging and important task in medicinal chemistry.
During the last few years, molecular hybridization represents a powerful tool in drug design, by merging two or more drug pharmacophores in a single hybrid multi-functional molecule. Usually, the resulting hybrid entity has superior properties compared with conventional classic drugs, with dual or multiple target mechanisms, better biological activity and specificity, less side effects and toxicity, less drug–drug interactions, etc. [2,3]. As a result of this approach, important advances have been achieved in antimicrobial therapy, some of the present drugs from the market have a hybrid structure (Figure 1) and some hybrid structures are in different clinical trials (Figure 2) [2,3,4,5,6,7,8].
A literature survey revealed that azines are privileged scaffolds in current medicinal chemistry and drug discovery, possessing a large variety of biological activities, such as: antibacterial, antifungal, antiplasmodial and antimalarial, anthelmintic, antitubercular, antiviral, anticancer, anti-inflammatory, antihypertensive, diuretic, antithrombic, anticoagulant, antidepressant, anxiolytic, anticonvulsant, analgesic, antiulcer, antidiabetic, antihistaminic, etc. [4,5,6,7,8]. As a matter of fact, the greatest majority of the existing drugs from the market contain in their structure a nitrogen heterocycle, some of them being a hybrid structure (Figure 1), which justifies the demand of the pharmaceutical industry for such drugs with nitrogen heterocycle skeleton.
Because of the above considerations, there is a large and urgent demand from the pharmaceutical industry for newer and better drugs with enhanced antimicrobial activity, with superior pharmacokinetic and pharmacodynamic properties, the hybrid drugs being a serious and preferential option.
In this review, we present an overview of the newest research concerning the synthesis and antimicrobial activity of hybrid azine derivatives. The main data reviewed in this paper are summarized in Table 1 presented below.

2. Results and Discussion

2.1. Six-Member Ring Azaheterocycles with One Nitrogen Atom. Hybrid Pyridine

In their attempt to identify new antimicrobial compounds, Eryılmaz et al. [9] designed and synthesized different hybrid pyridine derivatives bearing in the 2- and 4-position of the ring of a thiazole moiety. The synthesis was straight and efficient, involving a Hantzsch cyclocondensation of pyridine-2- and 4- carbothioamide 1 and 3 with acetophenone derivatives, when the desired hybrid 4-(R-2-yl)-2-(pyridin-2-yl)thiazole 2ae and 4-(R-2-yl)-2-(pyridin-4-yl)thiazole 4ae are obtained, Scheme 1. The synthesized compounds were tested for their antibacterial activity [four strains, Gram-positive (Bacillus cereus, Staphylococcus aureus) and Gram-negative (Escherichia coli, Pseudomonas aeruginosa)] and antifungal activity (one strain, Candida albicans) via minimal inhibitory concentration (MIC) method and DNA cleavage activity studies. The authors established interesting correlation structure-biological activity (SAR), the most relevant finding being that 4-pyridine thiazole hybrid compounds 4ae showed more potent activity than 2ae. The most promising compound was found to be 4c (MIC values 0.01 mM) exhibited on the bacterial strains Staphylococcus aureus and Bacillus cereus.
In a subsequent paper, some of the above authors (Cinarli et al. [10]) synthesized different hybrid aroylhydrazone-pyridine-metal derivatives. The newly hybrid aroylhydrazone-pyridine metal derivatives [ZnL2] 7 have been synthesized in two steps: an initial cyclocondensation of pyridine-2-acyl derivative 5 (with aroylhydrazone leading to pyridine-aroylhydrazone ligand 6) is followed by complexation with M2+ metal (Zn2+), Scheme 2.
The synthesized compounds were tested for their antibacterial activity (four strains, Pseudomonas aeruginosa, Escherichia coli, Bacillus cereus and Staphylococcus aureus) and antifungal (one strain, Candida albicans) activity via minimal inhibitory concentration method. The [ZnL2] 7 has been found to be more active than pyridine-aroylhydrazone ligand 6 in all microorganisms (MIC = 11.71 μg/mL for bacteria and MIC = 23.43 μg/mL for C. albicans). The authors claim that the synthesized new complex acts on microorganisms by disrupting the cell wall structure. The DNA binding interactions was also determined experimentally by spectrophotometric and electrochemical methods. The obtaining data indicate that ligand 6 and hybrid [ZnL2] 7 interact the most with guanine base, and charge transfer is from DNA guanine bases to the molecular structures. Moreover, antioxidant activity was determined, and the hybrid [ZnL2] 7 acted as a scavenger against peroxide radicals.
Trotsko et al. [11] designed and synthesized different hybrid pyridine derivatives bearing at the 2-, 3- or 4- position of the ring of a thiazolidine-2,4-dione moiety. The synthesis involve a condensation reaction of hydrazonyl-pyridine 8ac with the corresponding (2,4-dioxo-1,3-thiazolidin-5-yl/ylidene) 9a,b/10ac, which are leading to the desired hybrid pyridine-2,4-dioxo-1,3-thiazolidin-5-yl derivatives 11af or pyridine-2,4-dioxo-1,3-thiazolidin-5-ylidene derivatives 12ai, Scheme 3.
The in vitro antimycobacterial assay (Mycobacterium tuberculosis) of the newly obtained compounds reveals strong activity in the concentration range of 1–512 μg/mL and low cytotoxicity. Interesting SAR correlations have been performed, and the highest antimycobacterial activity (MIC = 1 μg/mL) was demonstrated for the hybrid pyridine derivatives bearing the thiazolidine-2,4-dione moiety at the 4-position of the pyridine ring (hybrids 11ac and 12gi).
Sanad et al. [12] have performed an interesting study concerning the in vitro antimicrobial activity of some newly hybrid thieno-pyrimidin-pyridine derivatives. The synthesized compounds belonged to different classes of substituted pyridine: thiophen-dihydropyridine 14, thiophen-pyrido-pyrimidin-4(1H)-one 15, and fused pyridine: pyrido-thiophen-triazolo-pyrimidine 16ac, thiophen-pyrido-thieno derivative 17, thiophen-pyrido-thieno-pyrimidin-4-one 18, thiophen-pyrido-thieno-pyrimidin-2,4-dione 19, thiophen-pyrido-thieno-pyrimidin-2-R-4-one 20, Scheme 4.
The synthetic approach is straight and efficient, involving typical organic chemistry reactions, mostly cyclocondensations. The synthesized compounds were tested in vitro for their antibacterial activity against Escherichia coli and Klebsiella pneumoniae as Gram-negative bacterial strains as well as against Staphylococcus aureus and Streptococcus mutans as Gram-positive bacterial strains. The obtained results (expressed as the diameter of inhibition zones (DIZ) and MIC) reveal that the thiophen-pyrido-thieno-pyrimidin-2-R-4-one 20a,b exhibit the strongest antibacterial activities against all the tested bacteria, in the range of 40–60 mm for inhibition zones, respectively, 4–16 μg/mL for MIC values.
Desai et al. [13] have studied the in vitro antimicrobial activities of some newly hybrid oxazino-pyridine derivatives. The desired compounds, oxazin-3(4H)-yl)phenyl)ethyildene)amino)-6-((arylidene)amino)-4-(4-chlorophenyl)-2-oxo-1,2-dihydropyridine 23aj, were synthesized in two steps, by cyclocondensation of oxazine 21 followed by condensation of the intermediate 22, Scheme 5.
The synthesized hybrid compounds were tested for their in vitro antibacterial activity against various bacteria (Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus, Streptococcus pyogenes) and fungus (Candida albicans, Aspergillus niger, Aspergillus clavatus) via the MIC method. Some compounds have proved to have a very powerful activity against bacteria E. coli (23h, MIC = 25 μg/mL) and against fungus C. albicans (23f, MIC = 50 μg/mL), respectively, A. clavatus (23h, MIC = 25 μg/mL).
Sribalan et al. [14] have studied thein vitroantimicrobial activity of some tetrazole-heterocycle hybrid derivatives. The synthesis supposes a cyclocondensation reaction of amide precursors 24 with sodium azide, when the corresponding tetrazolo-pyridine 25ad and tetrazolo-quinoline 26ae hybrids are obtained, Scheme 6.
The synthesized tetrazolo-pyridine 25ad and tetrazolo-quinoline 26ae hybrids were tested for their in vitro antibacterial activity against various bacteria (Klebsiella pneumoniae, Pseudomonas aeruginosa, Staphylococcus aureus, Streptococcus pyogenes) and the fungus Candida albicans. An interesting SAR correlation has been performed. The compound 25a (the pyridyl ring is decorated with n-butyl) proved to be the most active from the tetrazolo-pyridine series against all bacteria (DIZ in the range of 4–15 mm), having a superior inhibition to the standard drug (amikacin). The compound 26d (the quinoline ring is decorated with a piperidyl-sulfonamide moiety) proved to be the most active from the tetrazolo-quinoline series against all bacteria (DIZ in the range of 4–10 mm), having a comparable inhibition to the standard. The antifungal activity was negligible.
Kuthyala et al. [15] have studied the in vitro antimicrobial activity of some oxadiazolo-imidazopyridine hybrid derivatives. The synthesis was straight, involving a cyclocondensation reaction of hydrazonyl-imidazopyridine 27 with different benzoic acids, when the corresponding oxadiazolo-imidazopyridine hybrids 28aj were obtained, Scheme 7.
The synthesized oxadiazolo-imidazopyridine hybrids 28aj were tested for their in vitro antibacterial activity against various human bacterial pathogens (Escherichia coli, Klebsiella pneumoniae, Staphylococcus aureus, Bacillus subtilis) and the fungus Candida albicans and Aspergillus niger. An interesting SAR correlation has been performed. The compounds 28f and 28g have high activity against Gram-positive bacteria S. aureus (MIC = 3.12 μg/mL), while compound 28f proved to have high activity against fungus C. albicans (MIC = 12.5 μg/mL).
Ahirwaret al. [16] synthesized two new series of some 1,3,4-triazolo-pyridine hybrid derivatives and studied their antimicrobial activities. The synthesis was conducted in two steps: a cyclocondensation reaction of dithiocarbazate 29 with ammonia leading to the first class of hybrids triazolo-pyridine 30an, then an alkylation reaction of 30an with benzyl halide takes place leading to the second class of hybrids triazolo-pyridine 31an, Scheme 8.
The synthesized triazolo-pyridine hybrids 30an and 31an were evaluated for their in vitro antibacterial activity against Gram-positive bacteria (three strains: Staphylococcus aureus, Streptococcus pyogenes, Enterococcus faecalis) and Gram-negative bacteria (three strains: Escherichia coli, Pseudomonas aeruginosa, Acinetbacter baumannii) by MIC assay. From the tested compounds, two of them, 31h and 31i, have excellent activity against all strains (MIC in the range of 0.91–11 μg/mL).
Jaabil et al. [17] have studied the in vitro antimicrobial activities of some newly hybrid 1,2,3-triazolo-pyridine derivatives. The synthesis was green and efficient, under grinding strategy at room temperature, involving one-pot sequential multicomponent reactions of aryl aldehydes 32ar, malonitrile 33, methanol and 1,2,3-triazolyl ketone 34, when the corresponding 1,2,3-triazolyl-pyridine/cyanopyridine hybrids 35ar were obtained, Scheme 9.
The synthesized 1,2,3-triazolo-pyridine hybrids 35ar were screened for their in vitro antibacterial activity against three human bacterial strains, Staphylococcus aureus, Salmonella typhi and Escherichia coli, using the MIC method. Some of the 1,2,3-triazolyl cyanopyridine hybrids displayed a remarkable activity against the tested germs, better than tetracycline (standard drug), according to the R-substituent from the phenyl ring. The most active compounds were 35c (with R = −4-chloro-; MIC in the range of 50–90 μg/mL), 35e (with R = −2-methyl-; MIC in the range of 40–90 μg/mL) and 35r (with R = −2-thienyl; MIC in the range of 70–120 μg/mL). The hybrid 1,2,3-triazolo-pyridine compounds were also tested for their antioxidant activity in the assay by 2,2-diphenyl-1-picrylhydrazyl (DPPH) method, showing promising results.
Felefel et al. [18] synthesized three new series of some pyridine hybrid derivatives (namely pyrazole-pyridine 3741, triazolo-pyridine 4245 and triazino-pyridine 46) and studied their antimicrobial activities. The synthesis is using as starting material 6-(3,4-dimethylphenyl)-2-hydrazinyl-4-(thiophen-2-yl)-pyridine-3-carbonitrile 36 which react with different compounds with methylene active group (namely acetyl acetone, diethylmalonate, ethyl cyanoacetate, ethyl benzoylacetate and/or ethyl acetoacetate) to produce the desired pyrazole-pyridine hybrid derivatives 3741, Scheme 10.
The synthesis of triazolo-pyridines 4245 and tetrazolo-pyridines 46 use as starting material the same intermediate, the 6-(3,4-dimethylphenyl)-2-hydrazinyl-4-(thiophen-2-yl)-pyridine-3-carbonitrile 36, which react with the appropriate formic acid, acetic acid, benzoyl chloride, carbon disulfide, respectively, sodium nitrite, to produce the desired hybrid derivatives 4245 and 46, Scheme 11.
The synthesized pyridine hybrids 3746 were screened for their in vitro antibacterial activity against Gram-positive bacteria (Staphylococcus aureus and Bacillus subtilis), Gram-negative bacteria (Salmonella typhi and Escherichia coli) and fungus (Aspergillus flavus and Candida albicans) using the disk diffusion agar technique. Some of the hybrids have significant antimicrobial activity, the most active compounds being 37 with a DIZ in the range of 10–17 mm. The antioxidant activity was also tested.
Amperayani et al. [19] synthesized a library of piperine-pyridine hybrid derivatives and studied their antimicrobial activities. The reaction pathway is straight, in one step, involving an acylation reaction of various amino-pyridine derivatives 47ah, when the corresponding hybrids piperine-pyridine derivatives 48ah are obtained, Scheme 12.
The synthesized piperine-pyridine hybrid derivatives 48ah were tested for their in vitro antibacterial activity against some Gram-positive and Gram-negative bacterial strains (Bacillus subtilis, Streptobacillus, Staphylococcus aureus, Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, Enterococcus faecalis and Salmonella typhi) and fungus strains (Aspergillus niger, Aspergillus flavus, Aspergillus fumigatus and Candida albicans) using the disk diffusion agar technique. The piperine-pyridine hybrids 48a, 48d and 48h have very good activity against the Gram-negative strains E. coli, K. pneumoniae, E. faecalis and P. aeruginosa, having a DIZ in the range of 22–26 mm, superior to control standard drug). The antifungal activity of hybrids was moderate.

2.2. Six-Member Ring Azaheterocycles with One Nitrogen Atom. Hybrid Quinoline and Isoquinoline

In their attempt to obtain new quinoline derivatives with antimicrobial activity, Albayrak et al. [20] synthesized a library of 20 new triazolo-quinoline hybrid derivatives and studied their antimicrobial activities. The reaction pathway involves several steps (Scheme 13), starting from 8-nitroquinoline 53. The initial reduction reaction of 53 is leading to 8-aminoquinoline 54, which is suffering a subsequent N-alkylation with azido-iodo-propane 52a,b (generated from the corresponding bromo-alkyl alcohol) leading to alkyl-azide-quinolines 55 and 56. Finally, the alkyl-azide-quinoline derivatives are treated with the corresponding alkyne 57aj leading to the desired products, the triazolo-quinoline hybrid derivatives 58aj and 59aj.
The synthesized triazolo-quinoline hybrid derivatives 58aj and 59aj were tested for their in vitro antibacterial activity against some Gram-positive and Gram-negative bacterial strains (Bacillus subtilis, Streptococcus pneumoniae, Staphylococcus aureus, Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa and Enterococcus faecalis) and fungus strains (Candida parapsilosis and Candida albicans) using the disk diffusion agar technique. The triazolo-quinoline hybrid derivatives 58aj and 59aj manifest good activity against the tested strains. The most active compound was 58a, having excellent activity against E. coli, P. aeruginosa, K. pneumoniae, E. faecalis, S. aureus, S. pneumoniae, B. subtilis, C. albicans and C. parapsilosis. In some cases, the activity was several orders of magnitude superior to control drugs (DIZ of 58a was in the range of 35–250 mm; control, ampicillin, respectively, fluconazole have had a DIZ of 35 mm).
Hryhoriv et al. [21,22] synthesized two new classes of hybrid derivatives analogous to fluoroquinolones, namely piperidino-quinoline 61a,b and 1,2,3-triazolo-piperidino- quinoline 62ak, and studied their antimicrobial activities. The first class of hybrids was obtainedviaan N-alkylation reaction of piperidino-quinoline 60a,b, when the N-substituted-piperidino-quinoline hybrids 61a,b are obtained. A click cyclocondensation reaction of 61a,b occurs to the second class of hybrids, the 1,2,3-triazolo-piperidino-quinoline 62ak, Scheme 14.
The synthesized hybrid derivatives piperidino-quinoline 61a,b and 1,2,3-triazolo-piperidino-quinoline 62ak were tested for their in vitro antibacterial activity against standard bacterial strains Staphylococcus aureus and Escherichia coli, respectively, and the fungus Candida albicans using the disk diffusion agar technique. The antimicrobial assay was also made by some clinical bacterial strains S. aureus and E. coli, respectively, and fungus C. albicans using the same method. The hybrid, 1,2,3-triazolo-piperidino-quinoline 62c have a very good activity against the tested standard strains (DIZ in the range of 25–35 mm), having a superior inhibition zone to control (DIZ = 25 mm). Against clinical microbial strains, the activity was negligible.
Drweesh et al. [23] synthesized hybrid organic-inorganic derivatives and studied their antimicrobial activities, antiproliferative activity, and radical scavenging properties. In order to synthesize the desired palladium-quinoline derivatives 64ad, they used organic cation modulation, doing a complexation reaction with PdCl2 of the quinolines 63ad, Scheme 15.
The synthesized palladium-quinoline derivatives hybrids 64ad and the free ligands 63ad, were tested for their in vitro antimicrobial activity against 14 standard microbial strains (Gram-positive and Gram-negative bacteria, fungus: Bifidobacterium animalis, Lactobacillus plantarum, Bacillus subtilis, Staphylococcus aureus ATCC 663, Staphylococcus aureus ATCC 25923, Pseudomonas aeruginosa, Proteus mirabilis , Escherichia coli, Salmonella enterica, Candida albicans, Saccharomyces boulardii, Aspergillus flavus, Trichoderma viridae, Aspergillus niger). All hybrid compounds 64ad have high antimicrobial activity against all tested strains, with minimum inhibitory concentration values ranging from 1.95 to 250 μg/mL. The results of DNA interaction studies indicate that the hybrids 64ad and the free ligands 63ad, interact with the DNAvia an intercalation mechanism (the aromatic chromophore intercalates the base pairs of DNA; compound 64a has the highest binding affinity). The anticancer activity was also studied, with compounds 64a and 64b having selective and high cytotoxicity against human lung and breast cancer cells.
Nehra et al. [24] synthesized a series of triazole-benzothiazole-quinoline hybrids and studied their antimicrobial properties. The reaction pathway is straight and efficient (Scheme 16), involving a click cyclocondensation reaction of azido-alkyl-benzothiazole 65af (generated in situ from the corresponding bromo-alkyl derivative) with the corresponding alkyne-quinoline, leading to the desired products, triazole- -benzothiazole-quinoline hybrids 66af.
The synthesized hybrids 66af were evaluated for their in vitro antimicrobial activity against two Gram-positive strains (Staphylococcus aureus and Bacillus subtilis) and two Gram-negative strains (Escherichia coli and Pseudomonas aeruginosa) and two fungal strains (Candida tropicalis and Aspergillus terreus). The tested hybrids have good antimicrobial activity against both bacteria and fungus. The most promising compound was proved to be 66a, with an antibacterial (DIZ in the range of 15–17 mm) and antifungal (DIZ in the range of 21–34 mm) activity superior to reference ciprofloxacin (DIZ = 22 mm) and fluconazole (DIZ = 20 mm), respectively. Interesting molecular docking studies were also performed.
Awolade et al. [25] synthesized a library of triazole-quinoline hybrids and studied their antimicrobial properties. The reaction pathway is straight involving click chemistry of various azides with triple bond derivatives, via copper(I)-catalyzed azide-alkyne 3 + 2 dipolar cycloaddition reactions, Scheme 17.
The synthesized hybrids 67au, 68az, 69an and 70a,b were evaluated for their in vitro antimicrobial activity against ESKAPE microbial strains (bacteria and fungus: (Staphylococcus aureus, Escherichia coli, Acinetbacter baumannii, Klebsiella pneumoniae, Candida albicans and Candida neoformans). Some of the compounds proved to have a good and broad-spectrum of antibacterial activity, against methicillin-resistant S. aureus (MRSA), E. coli, A. baumannii, multidrug-resistant K. pneumoniae and the fungus C. albicans and C. neoformans (superior to control, fluconazole). The most promising antibacterial compound was proved to be 70b with an MIC = 75.39 μM against MRSA, E. coli, A. baumannii, and multidrug-resistant K. pneumoniae. The hybrid 70b also has a very good antifungal activity against C. albicans and C. neoformans with an MIC of 37.69 and 2.36 μM, respectively, superior to control fluconazole.
Ammar et al. [26] synthesized a series of thiazole-quinoline hybrids and studied their antimicrobial properties. In order to synthesize the desired compounds, they used the condensation reaction between formil-quinoline derivatives with amino-thiazole or sulfathiazole, when the desired Schiff’s base thiazole-quinoline 71 and 72, are obtained, Scheme 18.
Further, the condensation reaction between formil-quinoline derivatives with different thiazolone derivatives lead to hybrid thiazolone-quinoline derivatives 7376, Scheme 19.
Finally, the cyclization of different quinoline-thiosemicarbazone derivatives with the halogenated compounds lead to other hybrid thiazole-quinoline derivatives 7782, Scheme 20.
The synthesized hybrids 7182, were evaluated for theirin vitroantimicrobial activity against eight standard microbial strains, three Gram-positive bacteria (Staphylococcus aureus, Bacillus faecalis and Bacillus subtilis), three Gram-negative bacteria (Escherichia coli, Salmonella typhi and Pseudomonas aeruginosa), and two fungi (Candida albicans and Fusarium oxysporum). Some of the compounds have good antimicrobial activity, with MIC and MBC values ranging between 0.95 and 62.5 µg/mL, and 1.94 and 118.7 µg/mL, respectively. Two compounds, namely 77b and 73a, proved to be the most active of the series against S. aureus and E. coli having an MIC between 0.95 and 7.81 μg/mL, respectively a MBC between 3.31 and 15.62 μg/mL.
Using a similar strategy, some of the above authors (Eissa et al. [27]) synthesized a new series of thiazole-quinoline hybrids and studied their antimicrobial properties. In order to synthesize the desired compounds, they used the cyclization of quinoline-thiosemicarbazone derivatives with the halogenated compounds, when the corresponding hybrid thiazole-quinoline derivatives, 83af, 84af and 85af are obtained, Scheme 21.
The synthesized hybrids 83af, 84af and 85af, were evaluated for their in vitro antimicrobial activity against Gram-positive (five strains: Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus pyogenes, Bacillus subtilis and Enterococcus faecalis) and Gram-negative bacteria (five strains: Neisseria gonorrhoeae, Proteus vulgaris, Klebsiella pneumonia, Shigella flexneri and Pseudomonas aeruginosa), as well as fungus (five strains: Aspergillus fumigatus, Aspergillus clavatus, Candida albicans, Geotrichum candidum, and Penicillium marneffei). Some of the compounds displayed good antimicrobial activity, superior to the used control. The most active compound was found to be 85e, having a two-fold potency compared with gentamycin for inhibition of N. gonorrhoeae, four-fold potency compared with amphotericin B for the inhibition of A. fumigatus, equipotent activity compared with the reference drugs for inhibition of S. flexneri, S. pyogenes, P. vulgaris, A. clavatus, G. candidum and P. marneffei.
Lagdhir et al. [28] synthesized a library of piperazin-quinoline hybrids and studied their antimicrobial properties. The reaction pathway involves two steps (an alkylation and a condensation reaction), leading to the piperazin-quinoline hybrids 86al, Scheme 22.
The synthesized hybrids 86al were evaluated for their in vitro antibacterial (Staphylococcus aureus, Streptococcus pyogenes, Escherichia coli and Pseudomonas aeruginosa) and antifungal (Aspergillus clavatus, Aspergillus niger and Candida albicans) activity, antimalarial (Plasmodium falciparum) and antituberculosis (Mycobacterium tuberculosis) activity. Some of the compounds have good antibacterial and antifungal activity against S. aureus and C. albicans. The hybrids 86a, 86b, 86d, 86j and 86k, are the most active as an antimicrobial against S. aureus, having an MIC = 100 μg/mL, equal to the control drug ampicillin. The hybrid 86k has excellent antifungal activity against C. albicans, having an MIC = 250 μg/mL, two folds higher compared with the control drug griseofulvin. The antimalarial and antitubercular activity proved to be moderate for the majority of compounds.
Desai et al. [29] synthesized a series of pyridine-quinoline hybrids and evaluated it for their antimicrobial properties. The reaction pathway involves a cyclocondensation reaction of quinoline derivative with benzylidene-malononitril, when the corresponding pyridine-quinoline hybrids 87aj were obtained, Scheme 23.
The synthesized hybrids 87aj were evaluated for their in vitro antimicrobial activity against Gram-positive (two strains: Staphylococcus aureus and Staphylococcus pyogenes) and Gram-negative (two strains: Escherichia coli and Pseudomonas aeruginosa) bacteria, as well as to fungus (three strains: Aspergillus clavatus, Aspergillus niger and Candida albicans). Some of the compounds displayed promising antimicrobial activity. The hybrid 87i has the best antibacterial activity against E. coli, P. aeruginosa and S. aureus strains, with an MIC = 12.5 μg/mL, two folds higher compared with the control drug ciprofloxacin (MIC = 25 μg/mL). The most active compound against C. albicans was found to be 87e, having an MIC=25 μg/mL, much better compared with the control drug griseofulvin (MIC = 500 μg/mL).
Vishnuvardhan et al. [30] synthesized a library of triazole-quinoline hybrids and studied their antimicrobial properties. The reaction pathway involves a typical click cyclocondensation reaction of quinoline with a triple bond with aryl-azide derivatives, when the corresponding triazole-quinoline hybrids 88al, Scheme 24.
The synthesized hybrids 88al were evaluated for theirin vitroantimicrobial activity against Gram-positive (Staphylococcus aureus and Enterococcus faecalis) and Gram-negative (Escherichia coli and Pseudomonas aeruginosa) bacteria, as well as to fungus (Aspergillus niger and Candida albicans). Most of the hybrid compounds have good antimicrobial activity. The best antibacterial activity reveals the hybrids 88d, 88h and 88i, having a DIZ in the range of 16–21 mm, superior to control ampicillin (DIZ = 15 mm). The best antifungal activity reveals the hybrids 88d, 88h and 88k, having a DIZ in the range of 18–27 mm, superior to control griseofulvin (DIZ = 17 mm).
Abdel-Rahman et al. [31] synthesized a series of piperazin-quinoline hybrids derived from ciprofloxacin and studied their antimicrobial and anticancer properties. The reaction pathway involves the reaction of ciprofloxacin with the corresponding phenolic derivatives with an excess of formaldehyde, when the piperazin-quinoline hybrids 89aj are obtained, Scheme 25.
The synthesized hybrids 89aj were evaluated for their antimicrobial and anticancer activity. The antibacterial screening was preconformed on Gram-positive and Gram-negative strains: Staphylococcus aureus, MRSA clinical strain, MRSA reference strain, Escherichia coli and Pseudomonas aeruginosa. The obtained results reveal that the hybrid 89d has the best antibacterial activity against S. aureus, MRSA (reference strain) and MRSA (clinical strain) with an MIC of 0.57, 0.52, and 0.082 µg/mL, respectively, (compared with the reference standard drug ciprofloxacin which has an MIC of 1.63 µg/mL against S. aureus, an MIC of 1.45 µg/mL against MRSA reference, and an MIC of 0.84 µg/mL against MRSA clinical). The hybrid 89j exhibited the best antimicrobial activity against E. coli and P. aeruginosa, with an MIC of 0.036 and 0.043, respectively, (compared with the reference standard drug ciprofloxacin which has an MIC of 0.056 µg/mL against E. coli and an MIC of 1.27 µg/mL against P. aeruginosa).
Mohammed et al. [32] synthesized a series of glycosylated-quinoline hybrids derived from fluoroquinolone and studied their antimicrobial properties. The reaction pathway involves the reaction of ciprofloxacin with the corresponding phenolic derivative with an excess of formaldehyde, when the glycosylated-quinoline hybrids 9094 are obtained, Scheme 26.
The synthesized glycosylated-quinoline hybrids 9094 were evaluated for their antibacterial activity against various Gram-positive and Gram-negative bacteria: Escherichia coli, Listeria monocytogenes, Salmonella enterica, Pseudomonas aeruginosa, Listeria monocytogenes, E. coli clinical isolate (resistant to nalidixic acid, ciprofloxacin HCl and norfloxacin antibiotics), methicillin-resistant Staphylococcus aureus (MRSA), methicillin-sensitive Staphylococcus aureus (MSSA). The hybrids were also tested for their antifungal activity against fungi: Candida albicans, Aspergillus flavus, Fusarium solani, Stachybotrys chartarum and Penicillium chrysogenum. The hybrid compounds 90, 91 and 94a have excellent antimicrobial activity against a fluoroquinolone-resistant E. coli clinical isolate, comparable to controls ciprofloxacin and norfloxacin. The hybrid compound 91 also has good antifungal activity against C. albicans and P. chrysogenum.
Shruthi et al. [33] synthesized a series of piperazine-quinoline hybrids 95ae and morpholine-quinoline hybrids 96af and evaluate them for their antimicrobial properties. The reaction pathway is depicted in Scheme 27.
The synthesized hybrids 95ae and 96af were evaluated for their antibacterial (Acinetobacter baumanii, Enterococcus faecium, Klebsiella pneumonia, Pseudomonas aeruginosa, Escherichia coli and Staphylococcus aureus) and antitubercular (Mycobacterium tuberculosis) activity. Hybrid 95b has the best antibacterial activity against E. coli and S. aureus strains with an MIC of 4, respectively, 2 µg/mL, compared to standard drug vancomycin (MIC of 16, respectively, 0.5 µg/mL). Hybrids 95d, 95e and 96f exhibited the best antibacterial activity against A. baumaniistains with an MIC in the range of 1–2 µg/mL, compared to standard drug vancomycin (MIC = 0.5 µg/mL). Hybrids 95b, 95d and 95e also have promising antitubercular activity with an MIC of 4 µg/mL.
Kaur et al. [34] synthesized a series of 3- and 7- substituted-quinoline hybrids derived from fluoroquinolone and studied their antimicrobial properties. The reaction pathway involves the reaction of fluoroquinolone derivatives with the corresponding reagents, when the quinoline hybrids 97104a,b are obtained, Scheme 28 and Scheme 29.
The synthesized quinoline hybrids 97104a,b were evaluated for their antibacterial activity against four bacterial strains: Bacillus subtilis, Pseudomonas aeruginosa, Escherichia coli and Staphylococcus aureus. All hybrids 97104a,b have proved to be active against all bacterial strains, with an MIC value of 25 μg/mL which is fourfold more active compared to the standard drug ciprofloxacin (MIC = 100 μg/mL).
Insuasty et al. [35] synthesized a series of imidazolium-quinoline hybrids and studied their antimicrobial properties. The reaction pathway involves the reaction of 3-formyl-quinolone derivatives with the corresponding imidazolium salts, when the imidazolium-quinoline hybrids 105ah are obtained, Scheme 30.
The synthesized imidazolium-quinoline hybrids 105ah were evaluated for their antibacterial (Klebsiella pneumoniae, Escherichia coli and Staphylococcus aureus), antifungal (Cryptococcus neoformans) and antitubercular (Mycobacterium tuberculosis H37Rv and Mycobacterium bovis BCG) activities. Hybrid derivatives 105c,d demonstrated a remarkable antifungal activity against C. neoformans (MIC in the range of 15 µg/mL) while for the other fungal strains the activity is weak. The hybrids have modest antibacterial activity (both against Gram-positive and Gram-negative bacteria) as well as antitubercular activity.
Baartzes et al. [36] synthesized a series of benzimidazole-quinoline and ferrocenyl-quinoline hybrids and studied their antimicrobial properties. The reaction pathway involves the reaction of amino-quinoline derivatives with the corresponding formyl derivatives, when the benzimidazole-quinoline hybrids 106ae and ferrocenyl-quinoline hybrids 107ae are obtained, Scheme 31.
The synthesized quinoline hybrids 106ae and 107ae were evaluated for their antimalarial (Plasmodium falciparum and Plasmodium berghei) and antitubercular (Mycobacterium tuberculosis) activity. All hybrid derivatives are active against tested malaria strains and have modest activity against them. The most active hybrids against malarial strains have proved to be 106c and 107b, with an IC50 of 0.43, respectively, 0.32 µM, compared with the standard drug chlorquine (IC50 = 0.01 µM).
Fedorowicz et al. [37] synthesized a series of zwiterionic hybrids pyridine-fluoroquinolone 108ah and quinoline-fluoroquinolone 109ah and studied their antimicrobial properties. The reaction pathway involves a tandem Mannich-electrophilic amination reaction of isoxazolones derivatives and fluoroquinolone bearing a secondary amino group at position 7 of the quinoline ring, Scheme 32.
The synthesized quinoline hybrids 108ah and 109ah were evaluated for their antibacterial activity against Gram-positive and Gram-negative bacterial strains (laboratory and clinical: Staphylococcus aureus ATCC 6538, Staphylococcus aureus MRSA N315, Staphylococcus epidermidis ATCC 14990, Bacillus subtilis ATCC 6633, Escherichia coli ATCC 8739, Pseudomonas aeruginosa ATCC 9027, Proteus vulgaris NCTC 4635, Staphylococcus aureus MRSA 6347, Staphylococcus epidermidis MRSE 13199 and Serratia marcescens 12795) as well as for antibiofilm activity. The hybrid derivatives proved to have bactericidal and antibiofilm activity. The most active hybrids were found to be 109d and 109e, exhibiting good inhibition against all strains, with the IC50 values in the low micromolar range.
Borazjani et al. [38] synthesized a library of quinoline hybrids (benzothiazole-benzo-quinoline 110, imino-benzothiazole-benzo-quinoline 111ad, β-lactam-benzo-thiazole-benzo-quinoline 112am) and studied their antimicrobial properties. The reaction pathway involves a [2+2]-cycloaddition reaction of imines 111ad and ketenes derived from substituted acetic acids, Scheme 33.
The synthesized quinoline hybrids 110112 were evaluated for their antimicrobial activity against Gram-positive and Gram-negative bacterial strains: Staphylococcus aureus, Bacillus subtilis, Enterococcus faecalis, Salmonella typhi, Escherichia coli and Pseudomonas aeruginosa. From the β-lactam class, the assay indicates that the most active hybrids against E. coli and P. aeruginosa, are 112k and 112m, with an MIC of 42, respectively, 20 μg/mL, compared to standard drug gentamycin (MIC of 90, respectively, 5 μg/mL). From the imino-benzothiazole-benzo-quinoline class, the most active hybrids against P. aeruginosa and S. aureus, are 111ac, with an MIC of 42 μg/mL, compared to standard drug gentamycin (MIC of 5, respectively, 90 μg/mL).
Berry et al. [39] synthesized a series of peptide-fluoroquinolone hybrids and studied their antimicrobial properties. In order to synthesize the desired hybrids, the authors used solid-phase peptide synthesis, from levofloxacin fluoroquinolone with the corresponding peptide (oligopeptide), when the desired peptide-fluoroquinolone hybrids 113al are obtained, Scheme 34.
The synthesized peptide-fluoroquinolone hybrids 113al were evaluated for their antimicrobial activity against MDR bacterial strains, Gram-negative and Gram-positive: Pseudomonas aeruginosa, Escherichia coli, Klebsiella pneumoniae, Acinetobacter baumannii, methicillin-resistant Staphylococcus aureus (MRSA), methicillin-sensitive Staphylococcus aureus (MSSA), methicillin-resistant Staphylococcus epidermis (MRSE), Enterococcus faecalis, Enterobacter cloacae, Stenotrophomonas maltophilia. The assay indicates that all the peptide-hybrids have weak antibacterial activity. If the hybrids are mixed with fluoroquinolone (ciprofloxacin, levofloxacin and moxifloxacin) drugs, the resulting conjugates possess antimicrobial activity against MDR Gram-negative bacteria (clinical isolates, P. aeruginosa, E. coli, K. pneumoniae, A. baumannii), superior to reference levofloxacin.
Mermer et al. [40] synthesized a library of triazole- and oxadiazole-fluoroquinolone hybrids and studied their antimicrobial properties. The reaction pathway took placeviaseveral steps of sequential reactions, starting from phenyl piperazine. Finally, the corresponding triazole-fluoroquinolone 114a,b and oxadiazole-fluoroquinolone 115aj hybrids were obtained via a one-pot three-component Mannich reaction, Scheme 35. The reactions were performed both under conventional thermal heating and microwave, the last pathway being more advantageous.
The synthesized hybrids 114a,b and 115aj were tested for their antimicrobial activity (against Gram-positive and Gram-negative strains: Staphylococcus aureus, Enterococcus faecalis, Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, Acinetobacter haemolyticus), DNA gyrase and Topoisomerase IV inhibition potentials. The hybrids have good antimicrobial activity and displayed excellent DNA gyrase inhibition. The hybrids 114b, 115b and 115h exhibited the best antimicrobial activity against the tested strains. Thus, the hybrids have excellent activity against K. pneumoniae with an MIC of 0.25 µg/mL, compared with the standard drug gentamycin (MIC = 0.25 µg/mL). The hybrids also have excellent activity against A. haemolyticus and P. aeruginosa with an MIC in the range of 0.5–2 µg/mL, compared with the standard drug gentamycin (MIC = 0.78 µg/mL, respectively, MIC = 1.56 µg/mL). Against Gram-positive strain E. faecalis the hybrids have excellent activity with an MIC in the range of 0.5–8 µg/mL, compared with the standard drug ampicillin (MIC = 12.5 µg/mL).
Guo et al. [41] synthesized a library of oxadiazole-quinoline hybrids and studied their antibacterial properties. The reaction pathway is straight, involving an alkylation reaction of fluoroquinolone with the corresponding oxadiazole, when the desired oxadiazole-fluoroquinolone hybrids 116at were obtained, Scheme 36.
The synthesized oxadiazole-fluoroquinolone hybrids 116at were tested for their antibacterial activity against methicillin-resistant Staphylococcus aureus (MRSA) and laboratory Staphylococcus aureus. The hybrids displayed good antibacterial activity, one of the compounds 116k exhibited excellent antibacterial activity against both methicillin-resistant S. aureus and laboratory S. aureus, with an MIC in the range of 0.25–2 μg/mL, superior to control drug vancomycin (MIC = 2 μg/mL).
Wang et al. [42] synthesized a series of benzimidazole–quinoline hybrids and studied their antibacterial and antifungal properties. The reaction pathway involves an N-alkylation reaction of fluoroquinolone with the corresponding benzimidazole, when the desired benzimidazole-fluoroquinolone hybrids 117ag, 118a,b and 119af, were obtained, Scheme 37.
The synthesized benzimidazole-fluoroquinolone hybrids 117ag, 118a,b and 119af were screened against Gram-positive and Gram-negative bacteria, respectively, fungus (methicillin-resistant Staphylococcus aureus (MRSA), Enterococcus faecalis, Staphylococcus aureus, Staphylococcus aureus ATCC25923, Staphylococcus aureus ATCC29213, Klebsiella pneumonia, Escherichia coli, Pseudomonas aeruginosa, Acinetobacter baumanii, Pseudomonas aeruginosa ATCC27853, Escherichia coli ATCC25922, Candida albicans, Candida tropicalis, Aspergillus fumigatus, Candida albicans ATCC90023, Candida parapsilosis ATCC22019). The results of the assay were promising, with some hybrids having excellent antibacterial activity. The most active hybrids against K. pneumonia are 117a and 117c, with an MIC of 8 μg/mL, compared to the standard drug norfloxacin (MIC > 512 μg/mL).The most active hybrids against S. aureus are 119a and 119f, with an MIC of 4 μg/mL, compared to the standard drug norfloxacin (MIC = 64 μg/mL).
Bharadwaj et al. [43] synthesized a series of oxadiazole–quinoline hybrids and studied their antibacterial and antifungal properties. The reaction pathway involves a cyclocondensation reaction of hydrazinyl-quinoline derivative with the corresponding aromatic acids, when the desired oxadiazole–quinoline hybrids 120ag were obtained, Scheme 38.
The synthesized oxadiazole–quinoline hybrids 120ag were tested against clinical isolates Gram-positive and Gram-negative bacteria (Staphylococcus aureus, Bacillus cereus, Escherichia coli, Serratia marcescens), respectively, fungus (Aspergillus niger, Trichophyton mentagrophytes, Candida albicans, Candida parapsilosis). The antimicrobial activity of oxadiazole–quinoline derivatives was good, the hybrids 120a and 120f having the best antimicrobial activity against B. cereus with an MIC of 17, respectively, 24 μg/mL, compared to standard drug ampicilin (MIC = 16 μg/mL).
Tahaab et al. [44] synthesized a series of oxadiazole–quinoline hybrids and studied their leishmanicidal potential. The reaction pathway to obtain the oxadiazole–quinoline hybrids 121ar is depicted in Scheme 39.
The synthesized oxadiazole-quinoline hybrids 121ar were tested for their leishmanicidal activity against Leishmania major promastigote. Most of the synthesized hybrids have a good leishmanicidal activity, compound 121r was found to be the most active (IC50 = 0.10 μM) from the series, being 70 times more active than the standard drug (pentamidine, IC50 = 7 μM).
Irfan et al. [45] synthesized a series of triazole–quinoline hybrids and studied their antifungal properties. The reaction pathway involves a typical click cyclocondensation reaction of azide with a compound with a triple bond, when the desired triazole–quinoline hybrids 122ac were obtained, Scheme 40.
The synthesized triazole–quinoline hybrids 122ac were tested against fungus Candida albicans, both clinical isolates and laboratory strains [three FLC susceptible strains (C. albicans D27, C. albicans D31 and C. albicans D39) and one FLC resistant strain (C. albicans D15.9)]. The best antifungal activity was found for the hybrids 122a and 122b, having an MIC of 25 μg/mL for 122a and an MIC of 250 μg/mL for 122b, compared to control FLC (MIC >1 μg/mL)
Pandya et al. [46] synthesized a library of pyrazole–isoquinoline hybrids and studied their antimicrobial properties. The reaction pathway involves a palladium-catalyzed reaction of pyrazole derivatives with t-butyl-isocyanide, when the corresponding pyrazole–isoquinoline hybrids 123ag, were obtained, Scheme 41.
The synthesized pyrazole–isoquinoline hybrids 123ag were evaluated for their antimicrobial activity against different pathogenic strains: bacterial strains (Staphylococcus aureus, Escherichia coli, Enterococcus faecalis, Streptococcus pyogens and Vibrio cholera), fungal strains (Candida albicans, Candida glabrata, Candida krusei, Candida tropicalis and Candida parapsilosis) and tubercular strain (Mycobacterium tuberculosis). The antimicrobial activity of hybrids was very good, the hybrids 123e and 123g having the best antimicrobial activity, compared to standard drugs kanamycin and amphotericin B. Thus, the most active hybrids against S. aureus are 123e and 123g, having an MIC of 20 μM, respectively, 37 μM, compared to standard drug kanamycin (MIC of 31 μM). The most active hybrids against V. cholera are 123e and 123g, having an MIC of 41 μM, respectively, 90 μM, compared to the standard drug kanamycin (MIC of 62 μM). The hybrids 123e and 123g have the best antitubercular activity against M. tuberculosis with an MIC of 30 μg/mL, respectively, 32 μg/mL, compared to standard drugs rifampicin and isoniazide (MIC of 90 μg/mL).
Verma et al. [47] obtained a series of piperazine- and pyrimidine- isoquinoline hybrids and studied their antimicrobial properties. The piperazine-isoquinoline hybrids 126ah were synthesized by condensation of the carboxylic acid intermediates 124ad with appropriate aryl-piperazines, Scheme 38. The pyrimidine-isoquinoline hybrids 127ah were synthesized in two steps: an O-alkylation of the carboxylic acid intermediates 124ad (with ethylene dichloride), followed by an S-alkylation of the obtained compounds 125ad (with thio-pyrimidine), Scheme 42.
The synthesized piperazine- and pyrimidine-isoquinoline hybrids 124ah and 125ah were evaluated for their antibacterial and antifungal (Escherichia coli, Klebsiella pneumoniae, Staphylococcus aureus, Bacillus subtilis, Aspergillus niger, Aspergillus oryzae, Candida albicans and Pencillium chrysogenum), antioxidant, anticancer and antituberculosis (Mycobacterium tuberculosis) activities. The antibacterial assay indicates that three hybrids, namely 124a, 125a and 126e have the best activity against E. coli (with an MIC in the range of 1–3 μg/mL) and K. pneumoniae (with an MIC in the range of 1.5–3 μg/mL), compared with the standard drug ciprofloxacin (MIC = 1.5 μg/mL). The hybrids 125a, 126a and 127a also have excellentactivity against S. aureus (with an MIC in the range of 1–3 μg/mL) and B. subtilis (with an MIC in the range of 1.5–3 μg/mL), compared with the standard drug ciprofloxacin (MIC = 1.5 μg/mL, respectively, MIC = 3 μg/mL). The hybrids 125a, 126a and 127a have excellent activity against fungus A. niger, C. albicans, A. oryzae, and P. chrysogenum (with an MIC of 1.5 μg/mL), compared with the standard drug fluconazole (MIC = 1.5 μg/mL for A. niger and C. albicans, respectively, MIC = 3 μg/mL for A. oryzae, and P. chrysogenum). The hybrids 127b and 127e have the best activity against M. tuberculosis (MIC 1.0 mg/mL), compared with the standard drug rifampicin (MIC = 0.1mg/mL. The antioxidant and anticancer activity proved to be modest.

2.3. Our Group Recent Contributions

Our concern for obtaining new six-member ring azaheterocycle entities with antimicrobial activity for medicinal chemistry applications was started three decades ago [48,49,50,51] when we tried to obtain new diazines with good to excellent antibacterial and antifungal activities. Further, we will present some recent results obtained by us in the field of hybrid azines with antimicrobial activity.
In continuation of our concern for new compounds with antimicrobial activity, Diaconu et al. [52] synthesized a large library of hybrid imidazole- and benzimidazole-quinoline derivatives and studied their antimicrobial properties. The reaction pathway (Scheme 42) involves an initial N-acylation reaction of 8-aminoquinoline, followed by an N-alkylation of the -NH- amino group from imidazole/benzimidazole heterocycle, when the key imidazole-quinoline 128a,b and benzimidazole-quinoline 129a,b hybrids are obtained. Next, a quaternization reaction of N-imidazole atom with activated halogenated compounds leads to a second class of hybrids, the quaternary salts of imidazole- benzimidazole-quinolines, 130ak and 132ak (with one methylene group as linker) and 131ak and 133ak (with two methylene groups as linker). Finally, imidazolium and benzimidazolium ylides (generated in situ from the corresponding salts) react with dimethyl acetylenedicarboxylate (DMAD), generating another class of hybrid quinoline derivatives, the benzimidazole-quinoline cycloadducts 134ak, Scheme 43.
The synthesized hybrids were evaluated for their antimicrobial (Staphylococcus aureus, Escherichia coli and Candida albicans) and anticancer activities. The results of the antibacterial assay indicate that some hybrid compounds are biologically active in the range of nano-molar, five benzimidazole-quinoline hybrid salts (133c, 133d, 133f, 133h, 132h) have excellent activity against Gram-negative bacteria E. coli (DIZ in the range of 20–24 mm) superior to control gentamicin (DIZ of 12 mm) and one compound (131i) have excellent activity against Gram-positive bacteria S. aureus (DIZ of 20 mm) superior to control gentamicin (DIZ of 14 mm). The anticancer assay indicates that some benzimidazole-quinoline hybrid salts (133h, 132h, 133c, 133f) have excellent anticancer activity in the range of nano-molar, against some cancer cells (Leukemia, Breast cancer, Lung cancer and Ovarian cancer). Interesting SAR correlations have been performed.
In another research work, Diaconu et al. [53] synthesized a new series of hybrid imidazole- and benzimidazole-pyridine derivatives and studied their antimicrobial properties. The reaction pathway (Scheme 43) involves an N-acylation reaction of 2-aminopyridine, followed by an N-alkylation of the -NH- amino group from imidazole/benzimidazole heterocycle when the corresponding imidazole-pyridine 135 and benzimidazole-pyridine 136 hybrids are obtained. Finally, a quaternization reaction of N-imidazole atom with activated halogenated compounds leads to a second class of hybrids, the imidazole-pyridine 137a,b and benzimidazole-pyridine 138a,b salts, Scheme 44.
The synthesized hybrids were tested for their antimicrobial activities against Staphylococcus aureus, Escherichia coli and Candida albicans. The results of the antibacterial assay indicate that the imidazole- and benzimidazole-pyridine hybrids have interesting antimicrobial properties, especially the hybrid benzimidazole-pyridine salt 138a have a powerful antibacterial activity against Gram-positive strain S. aureus and Gram-negative germ E. coli (DIZ of 30 mm), superior to control drug gentamicin (DIZ of 18 mm).
Antoci et al. [54] synthesized a new series of hybrid bis(imidazole)- and bis(benzimidazole)-pyridine derivatives and studied their antimycobacterial activity. The reaction pathway (Scheme 44) involves an N-alkylation of the -NH- amino group from imidazole/benzimidazole heterocycle when the corresponding bis(imidazole)-pyridine 139, 140 and bis(benzimidazole)-pyridine 141 hybrids are obtained. In the next step, a quaternization reaction of N-imidazole atom with activated halogenated compounds leads to a second class of hybrids, the bis(imidazole)-pyridine 142ag and bis(benzimidazole)-pyridine 143ag salts, Scheme 45.
The synthesized hybrids were tested in a primary screening for their antimycobacterial activities against M. tuberculosis H37Rv under aerobic conditions, eight hybrids (namely 140, 141, 142e,f and 143c,eg) having excellent activity against Mtb H37Rv, with an MIC in the range of 17–92 μM. The most active antimycobacterial five compounds (namely 140, 141, 142f and 143c,f) were subjected to the secondary antimycobacterial assay. The obtained results indicate that our compounds are very active against both replicating and non-replicating Mtb (superior to control metronidazole), exhibited excellent intracellular activity, are active against drug-resistant Mtb strains, have no cytotoxicity, and three of them (142f and 143c,f) have a bactericidal mechanism of action. The results of ADMET pharmacokinetic experimental studies for hybrid 143f, reveal that this compound is truly a candidate for a future drug: a lower clearance rate, a great half-time in vivo, a low potential for drug–drug interactions with a high duration of action and lack of cytotoxicity. The best antitubercular activity has the hybrid 143e with an MIC of 17 μM, MBC of 50 μM, IC50 of 9 μM. Under anaerobic conditions (LORA) the hybrid 143e have the MIC of 120 μM and IC50 of 9 μM. Against resistant isolates of Mtb strains [five strains, INH-R1 and INH-R2 (strains resistant to isoniazid), RIF-R1 and RIF-R2 (strains resistant to rifampicin), FQ-R1 (strain resistant to fluoroquinolone)] the hybrid 143e have the MIC in the range of 10–30 μM and IC50 in the range of 10–20 μM. Against nontuberculous mycobacteria Mycobacterium avium and Mycobacterium abscessus, the hybrid 143e has the MIC in the range of 50–80 μM and IC50 in the range of 30–50 μM. The intracellular activity and cytotoxicity of the hybrid 143e were IC50 of 14 μM, respectively, and IC50 of 50 μM.
Furthermore, in a subsequent paper [55], some of the above authors performed a thorough molecular docking study in order to determine the binding sites and ADMET properties of the hybrid bis(imidazole)- and bis(benzimidazole)-pyridine derivatives. The obtained results indicate the most probable binding sites the G-quadruplex DNA string and DNA strain in complex with dioxygenase. The predicted ADMET properties are in accordance with the experimental one presented above [54].
Continuing our studies in the field of hybrid pyridine and quinoline derivatives, Mantu et al. [56] synthesized a new series of hybrid imidazole- and benzimidazole-pyridine and quinoline derivatives and studied their antimicrobial properties. The reaction pathway involves a quaternization reaction of N-imidazole atom with activated halogenated compounds, when the corresponding salts of imidazole- and benzimidazole-pyridine and quinoline hybrids are obtained, Scheme 46.
The synthesized hybrids were tested for their antimycobacterial and anticancer activities. The antimycobacterial assay reveals that our hybrids have modest activity against Mtb strains. The anticancer assay indicates that one of the hybrids, namely 129a, has a very good and selective antitumor activity against Renal Cancer A498 and Breast Cancer MDA-MB-468.
Diaconu et al. [57] synthesized two new series of hybrid quinoline-sulfonamide complexes and studied their antimicrobial activity. The reaction pathway involves a straight and efficient two-step procedure. In the first step, an acylation reaction of (3-, 4- or 8-)aminoquinoline derivatives with the corresponding benzenesulfonyl chlorides 148ac or quinolylsulfonyl chloride 148d took place, the desired ligands quinoline-sulfonamide type 149ad being obtained. In the second step, a complexation reaction of ligands 149ad with metal acetate (Cu2+, Co2+, Cd2+) or chloride (Zn2+) took place, with the desired hybrids quinoline-benzene-sulfonamide complexes (150ad, 151ad and 152ad) and quinoline-quinolinyl-sulfonamide complexes 153ad being obtained. The reaction pathway is depicted in Scheme 47 for the complexes derived from 8-aminoquinoline.
The synthesized hybrids were tested for their antimicrobial activity, with some of them having a very good antibacterial (Staphylococcus aureus, Escherichia coli) and antifungal (Candida albicans) activity. For instance, the hybrid N-(quinolin-8-yl)-4-chloro-benzenesulfonamide cadmium 153d has the best antibacterial activity, with a DIZ of 21 mm and an MIC of 19.04 × 10−5 mg/mL against S. aureus, a DIZ of 19 mm and an MIC of 609 × 10−5 mg/mL against E. coli, and an excellent antifungal activity against C. albicans, with a DIZ of 25 mm and an MIC of 19.04 × 10−5 mg/mL.
Al-Matarneh et al. [58] synthesized two new series of pyrrolo-quinoline and pyrrolo-isoquinoline hybrids and studied their antimicrobial activity. The reaction pathway involves a 3 + 2 dipolar cycloaddition reaction of the quinolinium and isoquinolinium ylides (generated in situ from the corresponding salts) with N-ethyl- or N-phenyl-maleimide, when the hybrid spyrrolo-quinoline 154ac and pyrrolo-isoquinoline 155ac are obtained, Scheme 48.
The synthesized hybrids pyrrolo-quinoline 154ac and pyrrolo-isoquinoline 155ac were tested for their antimicrobial activities but, unfortunately, the hybrids have no significant activity.
Danac et al. [59] synthesized a series of pyrrolo-phenanthroline hybrids and studied their antimycobacterial activity. The reaction pathway involves a 3 + 2 dipolar cycloaddition reaction of the phenanthrolinium ylides (generated in situ from the corresponding salts) with N-ethyl- or N-phenyl-maleimide, when the pyrrolo-phenanthroline hybrids 156ac are obtained, Scheme 49.
The synthesized hybrids 156ac were tested for their antimycobacterial activities. The antimycobacterial assay reveals that one hybrid, 156a, has a strong activity against the Mtb strain, with an IC50 of 56 μM.
Danac, Olaru et al. [60,61] synthesized a library of indolizine-pyridine hybrids and studied their antimycobacterial activity. The reaction pathway involves a 3 + 2 dipolar cycloaddition reaction of the 4,4′-bipyridinium mono-ylides (generated in situ from the corresponding salts) with ethyl propiolate, when the mono-indolizine-pyridine hybrids 157ae are obtained, Scheme 49. Next, a quaternization reaction of pyridine nitrogen atom with activated halogenated compounds 158al is leading to the salts of mono-indolizine-pyridine hybrids 159al. Finally, another 3 + 2 dipolar cycloaddition reaction with ethyl propiolate is leading to bis-indolizine-pyridine hybrids 160ad, Scheme 50.
The synthesized hybrids were tested in a primary screening for their antimycobacterial activities against M. tuberculosis H37Rv under aerobic conditions, the salts of mono-indolizine-pyridine hybrids 159al displaying an excellent activity against Mtb H37Rv, superior to the second-line antitubercular drugs cycloserine and pyrimethamine and, equal as the first line anti-TB Ethambutol. The most active antimycobacterial five compounds (namely 159a, 159c, 159d, 159h and 159i) were subjected to the secondary antimycobacterial assay (MIC, MBC, LORA, intracellular (macrophage) drug screening, and MTT cell proliferation). These mono-indolizine-pyridine hybrids have proved to be very active against replicating and non-replicating M. tuberculosis, are active against both extracellular and intracellular organisms, have a bactericidal mechanism of action, and had basically no toxicity. The best antitubercular activity has the hybrid 159i with an MIC of 8 μM, MBC of 3 μM, IC50 of 7 μM. Under anaerobic conditions (LORA) the hybrid 159i have the MIC of 63 μM and IC50 of 1.9 μM. Against resistant isolates of Mtb strains [five strains, INH-R1 and INH-R2 (strains resistant to isoniazid), RIF-R1 and RIF-R2 (strains resistant to rifampicin), FQ-R1 (strain resistant to fluoroquinolone)] the hybrid 159i have the MIC in the range of 6–22 μM and IC50 in the range of 6–12 μM. Against nontuberculous mycobacteria Mycobacterium avium and Mycobacterium abscessus, the hybrid 159i has the MIC in the range of 23–50 μM and IC50 in the range of 14–18 μM. The intracellular activity and cytotoxicity of the hybrid 159i were IC50 of 5 μM, respectively IC50 of 2 μM.

3. Perspectives and Conclusions

To conclude, we report herein the latest recent advances concerning the synthesis and antimicrobial properties of hybrid azine derivatives. The literature data presented in this review indicate that there is a great urgency insociety and pharmaceutical industry to develop new antimicrobial drugs for the treatment of infectious diseases. Moreover, the data indicate that a modern approach used to overcome the drawbacks of infectious diseases is to use molecular hybridization strategy, as a new modern approach in drug discovery. The hybrid pyridine, quinoline, isoquinoline and their fused derivatives have invaluable importance in modern antimicrobial therapy, the results presented in this review indicate that they have a large variety of antimicrobial activities, including antibacterial, antifungal, antimycobacterial, antileishmanial, antimalarial, antiviral, etc.
We show that the best methods for the synthesis of hybrid compounds are cyclocondensation, condensation and simple typical organic chemistry reactions such as alkylation, acylation, etc. We also show that many hybrid compounds have excellent antimicrobial activity, the combination of an azine moiety with a five-member ring azaheterocycle being the best approach to obtain drugs with improved and superior antimicrobial properties. A special mention has to be made about the obtained results in antituberculosis therapy, where the use of hybrids with pyridine or by pyridine merged with an imidazole or benzimidazole moiety seems to be a very efficient approach in treatment, in some cases, the hybrids had a spectacular antitubercular activity, including a bactericidal mechanism of action. Moreover, the fact that some of these hybrids are in different clinical trials is a good and solid argument for further research in this field.
Finally, having in view the above consideration, we encourage and underline further studies in the field of hybrid azine merged with a five-member ring azaheterocycle, which appears to be the most promising field of research within this area.

Author Contributions

Design, conception and writing were performed by V.M. and I.I.M.; D.A.-M., I.B. and A.A. were involved in conception of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant of the Romanian Ministry of Education and Research, CNCS—UEFISCDI, project number PN-III-P4-ID-PCE-2020-0371, within PNCDI III.

Data Availability Statement

Not applicable.

Acknowledgments

The authors are thankful to Romanian Ministry of Research, Innovation and Digitization, within Program 1—Development of the national RD system, Subprogram 1.2—Institutional Performance—RDI excellence funding projects, Contract no.11PFE/30.12.2021 and project POC/448/1/1 Research Centre with Integrated Techniques for Atmospheric Aerosol Investigation in Romania-RECENT AIR (grant agreement MySMIS no. 127324) and CERNESIM Center, within the Institute for Interdisciplinary Research at the Alexandru Ioan Cuza University of Iasi, for infrastructure used.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADMETAbsorption, Distribution, Metabolism, Excretion, Toxicity
ESKAPEan acronym for the six highly virulent and antibiotic- resistant bacterial pathogens: Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp.
MICminimal inhibitory concentration
MBCminimal bactericidal concentration
MRSAmethicillin-resistant Staphylococcus aureus
DIZdiameter of inhibition zones
DNAdesoxyribonucleic acid
SARstructure activity relationship
IC50inhibitory concentration at 50%
LORALow oxygen recovery assay
INH-R1 and INH-R2strains resistant to isoniazid
RIF-R1 and RIF-R2strains resistant to rifampicin
FQ-R1strain resistant to fluoroquinolone
NTMnontuberculous mycobacteria
O-alkylationoxigen-alkylation
N-alkylationnitrogen-alkylation
S-alkylationsulphur-alkylation
ttert (eg, t—butyl means tert-butyl)
iiso (eg, i—propyl means iso-propyl)
nnormal (eg, n—butyl means normal-butyl)
o, m, portho, meta, para
Memethyl
Etethyl
Prpropyl
Bubutyl
Phphenyl
OMemethoxy
Bacillus cereusB. cereus
Bifidobacterium animalisB. animalis
Bacillus subtilisB. subtilis
Enterococcus faecalisE. faecalis
Lactobacillus plantarumL. plantarum
Staphylococcus aureusS. aureus
Staphylococcus epidermidisS. epidermidis
Streptococcus pneumoniaeS. pneumoniae
Streptococcus mutansS. mutans
Streptococcus pyogenesS. pyogenes
Acinetobacter baumanniiA. baumannii
Escherichia coliE. coli
Klebsiella pneumoniaeK. pneumoniae
Neisseria gonorrhoeaeN. gonorrhoeae
Proteus mirabilisP. mirabilis
Proteus vulgarisP. vulgaris
Pseudomonas aeruginosaP. aeruginosa
Salmonella entericaS. enterica
Salmonella typhiS. typhi
Shigella flexneriS. flexneri
Aspergillus clavatusA. clavatus
Aspergillus nigerA. niger
Aspergillus flavusA. flavus
Aspergillus fumigatusA. fumigatus
Candida albicansC. albicans
Candida parapsilosisC. parapsilosis
Candida parapsilosisC. parapsilosis
Cryptococcus neoformansC. neoformans
Geotrichum candidumG. candidum
Penicillium marneffeiP. marneffei
Saccharomyces boulardiiS. boulardii
Trichoderma viridaeT. viridae
Mycobacterium tuberculosisM. tuberculosis, Mtb
Mycobacterium aviumM. avium
Mycobacterium abscessusM. abscessus
WHOWorld Health Organization

References

  1. WHO. Global Action Plan on Antimicrobial Resistance: WHO. 2016. Available online: https://www.who.int/publications/i/item/9789241509763 (accessed on 12 April 2022).
  2. Bansal, Y.; Silakari, O. Multifunctional compounds: Smart molecules for multifactorial diseases. Eur. J. Med. Chem. 2014, 76, 31–42. [Google Scholar] [CrossRef] [PubMed]
  3. Bremner, J.B.; Ambrus, J.I.; Samosorn, S. Dual action-based approaches to antibacterial agents. Curr. Med. Chem. 2007, 14, 1459–1477. [Google Scholar] [CrossRef] [PubMed]
  4. Matada, B.S.; Pattanashettar, R.; Yernale, N.G. A comprehensive review on the biological interest of quinoline and its derivatives. Bioorg. Med. Chem. 2021, 32, 115973. [Google Scholar] [CrossRef]
  5. Chourasiya, S.S.; Kathuria, D.; Wani, A.A.; Bharatam, P.V. Azines: Synthesis, structure, electronic structure and their applications. Org. Biomol. Chem. 2019, 17, 8486–8521. [Google Scholar] [CrossRef] [PubMed]
  6. Amariucai-Mantu, D.; Antoci, V.; Sardaru, M.C.; Al Matarneh, C.M.; Mangalagiu, I.I.; Danac, R. Fused pyrrolo-pyridines and pyrrolo-(iso) quinoline as anticancer agents. Phys. Sci. Rev. 2022, 1. [Google Scholar] [CrossRef]
  7. Amariucai-Mantu, D.; Mangalagiu, V.; Danac, R.; Mangalagiu, I.I. Microwave assisted reactions of azaheterocycles for medicinal chemistry applications. Molecules 2020, 25, 716. [Google Scholar] [CrossRef]
  8. Luca, M.C.; Tura, V.; Mangalagiu, I.I. Considerations concerning design and mechanism of action of a new class of dual DNA intercalators. Med. Hypotheses 2010, 75, 627–629. [Google Scholar] [CrossRef]
  9. Eryılmaz, S.; Turkcelikoglu, E.; Idil, O.; Inkaya, I.; Kozak, Z.; Mısır, E.; Gul, M. Derivatives of pyridine and thiazole hybrid: Synthesis, DFT, biological evaluation via antimicrobial and DNA cleavage activity. Bioorg. Chem. 2020, 95, 103476. [Google Scholar] [CrossRef]
  10. Cinarli, M.; Ataol, C.Y.; Cinarli, E.; Idil, O. Synthesis, characterization, biological, X-ray diffraction analysis andcomputational chemistry studies of new 2-acetylpyridine derivativehydrazone and its Zn(II) complex. J. Mol. Struct. 2020, 1213, 128152. [Google Scholar] [CrossRef]
  11. Trotsko, N.; Golus, J.; Kazimierczak, P.; Paneth, A.; Przekora, A.; Ginalska, G.; Wujec, M. Synthesis and antimycobacterial activity of thiazolidine-2,4-dione based derivatives with halogenbenzohydrazones and pyridinecarbohydrazones substituents. Eur. J. Med. Chem. 2020, 189, 112045. [Google Scholar] [CrossRef]
  12. Sanad, S.M.H.; Ahmed, A.A.M.; Mekky, A.E.M. Efficient synthesis and molecular docking of novel antibacterial pyrimidines and their related fused heterocyclic derivatives. J. Heterocycl.Chem. 2020, 57, 590–605. [Google Scholar] [CrossRef]
  13. Desai, N.C.; Bhatt, N.B.; Joshi, S.B.; Jadeja, K.A.; Khedkar, V.M. Synthesis, Antimicrobial Activity and 3D-QSAR Study of Hybrid Oxazine Clubbed Pyridine Scaffolds. ChemistrySelect 2019, 4, 7541–7550. [Google Scholar] [CrossRef]
  14. Sribalan, R.; Banuppriya, G.; Kirubavathi, M.; Padmini, V. Synthesis, biological evaluation and in silico studies of tetrazole-heterocycle hybrids. J. Mol. Struct. 2019, 1175, 577–586. [Google Scholar] [CrossRef]
  15. Kuthyala, S.; Shankar, M.K.; Nagaraja, G.K. Synthesis, Single-Crystal X-ray, Hirshfeld and Antimicrobial Evaluation of some New Imidazopyridine Nucleus Incorporated with Oxadiazole Scaffold. ChemistrySelect 2018, 3, 12894–12899. [Google Scholar] [CrossRef]
  16. Ahirwar, J.; Ahirwar, D.; Lanjhiyana, S.; Jha, A.K.; Dewangan, D.; Badwaik, H. Synthesis, Characterization, Molecular Modeling, and Biological Evaluation of 1,2,4-Triazole-pyridine Hybrids as Potential Antimicrobial Agents. J. Heterocycl. Chem. 2018, 55, 2598–2609. [Google Scholar] [CrossRef]
  17. Jaabil, G.; Ranganathan, R.; Ponnuswamy, A.; Suresh, P.; Shanmugaiah, V.; Ravikumar, C.; Murugavel, S.A. Green and Efficient Synthesis of Bioactive 1, 2, 3-Triazolyl-Pyridine/Cyanopyridine Hybrids via One-Pot Multicomponent Grinding Protocol. ChemistrySelect 2018, 3, 10388–10393. [Google Scholar] [CrossRef]
  18. Flefel, E.M.; El-Sofany, W.I.; El-Shahat, M.; Naqvi, A.; Assirey, E. Synthesis, molecular docking and in vitro screening of some newly synthesized triazolopyridine, pyridotriazine and pyridine-pyrazole hybrid derivatives. Molecules 2018, 23, 2548. [Google Scholar] [CrossRef]
  19. Amperayani, K.R.; Kumar, K.N.; Parimi, U.D. Synthesis and in vitro and in silico antimicrobial studies of novel piperine–pyridine analogs. Res. Chem. Intermed. 2018, 44, 3549–3564. [Google Scholar] [CrossRef]
  20. Albayrak, F.; Çiçek, M.; Alkaya, D.; Kulu, I. Design, synthesis and biological evaluation of 8-aminoquinoline-1,2,3-triazole hybrid derivatives as potential antimicrobial agents. Med. Chem. Res. 2022, 31, 625–665. [Google Scholar] [CrossRef]
  21. Hryhoriv, H.; Mariutsa, I.; Kovalenko, S.M.; Georgiyants, V.; Perekhoda, L.; Filimonova, N.; Geyderikh, O.; Sidorenko, L. The Search for New Antibacterial Agents among 1,2,3-Triazole Functionalized Ciprofloxacin and Norfloxacin Hybrids: Synthesis, Docking Studies, and Biological Activity Evaluation. Sci. Pharm. 2022, 90, 2. [Google Scholar] [CrossRef]
  22. Hryhoriv, H.; Mariutsa, I.; Kovalenko, S.M.; Sidorenko, L.; Perekhoda, L.; Filimonova, N.; Geyderikh, O.; Georgiyants, V. Structural modification of ciprofloxacin and norfloxacin for searching new antibiotics to combat drug-resistant bacteria. Sci. Pharm. Sci. 2021, 5, 4–11. [Google Scholar]
  23. Drweesh, E.A.; Kucharova, V.; Volarevic, V.; Miloradovic, D.; Ilic, A.; Radojevic, I.D.; Rakovic, I.R.; Smolkova, R.; Vilkova, M.; Sabolov, D.; et al. Low-dimensional compounds containing bioactive ligands. Part XVII: Synthesis, structural, spectral and biological properties of hybrid organic-inorganic complexes based on [PdCl4]2− with derivatives of 8-hydroxyquinolinium. J. Inorg. Biochem. 2022, 228, 111697. [Google Scholar] [CrossRef] [PubMed]
  24. Nehra, N.; Tittal, R.K.; Ghule, V.D. 1,2,3-Triazoles of 8-Hydroxyquinoline and HBT: Synthesis and Studies (DNA Binding, Antimicrobial, Molecular Docking, ADME, and DFT). ACS Omega 2021, 6, 27089–27100. [Google Scholar] [CrossRef] [PubMed]
  25. Awolade, P.; Cele, N.; Kerru, N.; Singh, P. Synthesis, antimicrobial evaluation, and in silico studies of quinoline—1H-1,2,3-triazole molecular hybrids. Mol. Divers. 2021, 25, 2201–2218. [Google Scholar] [CrossRef]
  26. Ammar, Y.A.; El-Hafez, S.M.A.A.; Hessein, S.A.; Ali, A.M.; Askar, A.A.; Ragab, A. One-pot strategy for thiazole tethered 7-ethoxy quinoline hybrids: Synthesis and potential antimicrobial agents as dihydrofolate reductase (DHFR) inhibitors with molecular docking study. J. Mol. Struct. 2021, 1242, 130748. [Google Scholar] [CrossRef]
  27. Eissa, S.I.; Farrag, A.M.; Abbas, S.Y.; El Shehry, M.F.; Ragab, A.; Fayed, E.A.; Ammar, Y.A. Novel structural hybrids of quinoline and thiazole moieties: Synthesis and evaluation of antibacterial and antifungal activities with molecular modeling studies. Bioorg. Chem. 2021, 110, 104803. [Google Scholar] [CrossRef]
  28. Lagdhir, M.; Pandya, C.; Pandya, A.; Vekariya, R.H.; Rajani, D.P. Design and synthesis of new quinoline hybrid derivatives and their antimicrobial, antimalarial and antitubercular activities. Indian J. Chem. Sect. B 2021, 60, 986–998. [Google Scholar]
  29. Desai, N.C.; Harsora, J.P.; Mehta, H.K. 2-Pyridone quinoline hybrids as potent antibacterial and antifungal agents. Indian J. Chem. Sect. B 2021, 60, 261–266. [Google Scholar]
  30. Vishnuvardhan, M.; Pradeep, M.; Gangadhar, T. An efficient microwave assisted synthesis and antimicrobial activity of novel p-Tolyloxyquinoline-Triazole hybrid derivatives. Chem. Data Collect. 2021, 31, 100612. [Google Scholar] [CrossRef]
  31. Abdel-Rahman, I.M.; Mustafa, M.; Mohamed, S.A.; Yahia, R.; Abdel-Aziz, M.; Abuo-Rahma, G.; Hayallah, A.M. Novel Mannich bases of ciprofloxacin with improved physicochemical properties, antibacterial, anticancer activities and caspase-3 mediated apoptosis. Bioorg. Chem. 2021, 107, 104629. [Google Scholar] [CrossRef]
  32. Mohammed, A.A.M.; Suaifan, G.A.R.Y.; Shehadeh, M.B.; Okechukwu, P.N. Design, synthesis and antimicrobial evaluation of novel glycosylated-fluoroquinolones derivatives. Eur. J. Med. Chem. 2020, 202, 112513. [Google Scholar] [CrossRef] [PubMed]
  33. Shruthi, T.G.; Subramanian, S.; Eswaran, S. Design, synthesis and study of antibacterial and antitubercular activity of quinoline hydrazone hybrids. Heterocycl. Commun. 2020, 26, 137–147. [Google Scholar]
  34. Kaur, G.; Kaur, M.; Sharad, L.; Bansal, M. Theoretical molecular predictions and antimicrobial activities of newly synthesized molecular hybrids of norfloxacin and ciprofloxacin. J. Heterocycl. Chem. 2020, 57, 225–237. [Google Scholar] [CrossRef]
  35. Insuasty, D.; Vidal, O.; Bernal, A.; Marquez, E.; Guzman, J.; Insuasty, B.; Quiroga, J.; Svetaz, L.; Zacchino, S.; Puerto, G.; et al. Antimicrobial activity of quinoline-based hydroxyimidazolium hybrids. Antibiotics 2019, 8, 239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Baartzes, N.; Stringer, T.; Seldon, R.; Warner, D.F.; Taylor, D.; Wittlin, S.; Chibale, K.; Smith, G.S. Bioisosteric ferrocenyl aminoquinoline-benzimidazole hybrids: Antimicrobial evaluation and mechanistic insights. Eur. J. Med. Chem. 2019, 180, 121–133. [Google Scholar] [CrossRef] [PubMed]
  37. Fedorowicz, J.; Sączewski, J.; Konopacka, A.; Waleron, K.; Lejnowski, D.; Ciura, K.; Toma, T.; Skok, Z.; Savijoki, K.; Morawska, M.; et al. Synthesis and biological evaluation of hybrid quinolone-based quaternary ammonium antibacterial agents. Eur. J. Med. Chem. 2019, 179, 576–590. [Google Scholar] [CrossRef]
  38. Borazjani, N.; Jarrahpour, A.; Rad, J.A.; Mohkam, M.; Behzadi, M.; Ghasemi, Y.; Mirzaeinia, S.; Karbalaei-Heidari, H.R.; Ghanbari, M.M.; Batta, G.; et al. Design, synthesis and biological evaluation of some novel diastereoselective β-lactams bearing 2-mercaptobenzothiazole and benzoquinoline. Med. Chem. Res. 2019, 28, 329–339. [Google Scholar] [CrossRef]
  39. Berry, L.; Domalaon, R.; Brizuela, M.; Zhanel, G.G.; Schweizer, F. Polybasic peptide-levofloxacin conjugates potentiate fluoroquinolones and other classes of antibiotics against multidrug-resistant Gram-negative bacteria. MedChemComm 2019, 10, 517–527. [Google Scholar] [CrossRef]
  40. Mermer, M.; Faiz, O.; Demirbas, A.; Demirbas, N.; Alagumuthu, M.; Arumugam, V. Piperazine-azole-fluoroquinolone hybrids: Conventional and microwave irradiated synthesis, biological activity screening and molecular docking studies. Bioorg. Chem. 2019, 85, 308–318. [Google Scholar] [CrossRef]
  41. Guo, Y.; Xu, T.; Bao, C.; Liu, Z.; Fan, J.; Yang, R.; Qin, S. Design and synthesis of new norfloxacin-1,3,4-oxadiazole hybrids as antibacterial agents against methicillin-resistant Staphylococcus aureus (MRSA). Eur. J. Pharm. Sci. 2019, 136, 104966. [Google Scholar] [CrossRef]
  42. Wang, Y.N.; Bheemanaboina, R.R.Y.; Gao, W.W.; Cang, J.; Cai, G.X.; Zhou, C.H. Discovery of Benzimidazole–Quinolone Hybrids as New Cleaving Agents toward Drug-Resistant Pseudomonas aeruginosa DNA. ChemMedChem 2018, 13, 1004–1017. [Google Scholar] [CrossRef] [PubMed]
  43. Bharadwaj, S.S.; Poojary, B.; Kumar, M.S.; Byrappa, K.; Nagananda, K.S.; Chaitanya, A.K.; Zaveri, K.; Yarla, N.S.; Shiralgi, Y.; Kudva, A.K.; et al. Design, synthesis and pharmacological studies of some new quinoline Schiff bases and 2,5-(disubstituted-[1,3,4])-oxadiazoles. New J. Chem. 2017, 41, 8568–8585. [Google Scholar] [CrossRef]
  44. Tahaab, M.; Ismailab, N.H.; Alic, M.; Rashidc, U.; Imranab, S.; Uddind, N.; Khane, K.M. Molecular hybridization conceded exceptionally potent quinolinyl-oxadiazole hybrids through phenyl linked thiosemicarbazide antileishmanial scaffolds: In silico validation and SAR studies. Bioorg. Chem. 2017, 71, 192–200. [Google Scholar]
  45. Irfan, M.; Alam, S.; Manzoor, N.; Abid, M. Effect of quinoline based 1,2,3-triazole and its structural analogues on growth and virulence attributes of Candida albicans. PLoS ONE 2017, 12, e0175710. [Google Scholar] [CrossRef]
  46. Pandya, K.M.; Battula, S.; Naik, P.J. Pd-catalyzed post-Ugi intramolecular cyclization to the synthesis of isoquinolone-pyrazole hybrid pharmacophores & discover their antimicrobial and DFT studies. Tetrahedron Lett. 2021, 81, 153353. [Google Scholar]
  47. Verma, V.A.; Saundane, A.R.; Meti, R.S.; Vennapu, D.R. Synthesis of novel indolo[3,2-c]isoquinoline derivatives bearing pyrimidine, piperazine rings and their biological evaluation and docking studies against COVID-19 virus main protease. J. Mol. Struct. 2021, 1229, 129829. [Google Scholar] [CrossRef]
  48. Ungureanu, M.; Mangalagiu, I.I.; Grosu, G.; Petrovanu, M. Antimicrobial activity of some new pyridazine derivatives. Ann. Pharm. Fr. 1997, 55, 69–72. [Google Scholar]
  49. Mangalagiu, I.I.; Ungureanu, M.; Mangalagiu, G.; Grosu, G.; Petrovanu, M. Antimicrobial activity of some pyrimidinium compounds. Ann. Pharm. Fr. 1998, 56, 181–183. [Google Scholar]
  50. Mangalagiu, G.; Ungureanu, M.; Grosu, G.; Mangalagiu, I.I.; Petrovanu, M. New pyrrolo-pyrimidine derivatives with antifungal or antibacterial properties in vitro. Ann. Pharm. Fr. 2001, 59, 139–140. [Google Scholar]
  51. Moldoveanu, C.; Mangalagiu, G.; Drochioiu, G.; Caprosu, M.; Petrovanu, M.; Mangalagiu, I.I. New Antituberculosis Compounds Derived from Diazine. An. Stiint. Univ. “Al. I. Cuza” Iasi Chem. 2003, 11, 367–374. [Google Scholar]
  52. Diaconu, D.; Antoci, V.; Mangalagiu, V.; Amariucai-Mantu, D.; Mangalagiu, I.I. Quinoline-imidazole/benzimidazole derivatives as dual- / multi- targeting hybrids inhibitors with anticancer and antimicrobial activity. Sci. Rep. 2022, in press. [Google Scholar]
  53. Diaconu, D.; Amariucai-Mantu, D.; Antoci, V.; Ciorteanu, R.; Mangalagiu, V.; Mangalagiu, I.I. Design and synthesis of new hybrid pyridine imidazole/benzimidazole salts with antibacterial activity. Rev. Roum. Chim. 2022, 67, 89–92. [Google Scholar]
  54. Antoci, V.; Cucu, D.; Zbancioc, G.; Moldoveanu, C.; Mangalagiu, V.; Amariucai-Mantu, D.; Aricu, A.; Mangalagiu, I.I. Bis-(imidazole/benzimidazole)-pyridine derivatives: Synthesis, structure and antimycobacterial activity. Part XII. Future Med. Chem. 2020, 12, 207–222. [Google Scholar] [CrossRef] [PubMed]
  55. Lungu, C.N.; Bratanovici, B.I.; Grigore, M.M.; Antoci, V.; Mangalagiu, I.I. Hybrid Imidazole-Pyridine Derivatives: An Approach to Novel Anticancer DNA Intercalators. Curr. Med. Chem. 2020, 27, 154–169. [Google Scholar] [CrossRef]
  56. Mantu, D.; Antoci, V.; Moldoveanu, C.; Zbancioc, G.; Mangalagiu, I.I. Hybrid imidazole (benzimidazole)/pyridine (quinoline) derivatives and evaluation of their anticancer and antimycobacterial activity. J. Enzym. Inhib. Med. Chem. 2016, 31, 96–103. [Google Scholar] [CrossRef]
  57. Diaconu, D.; Mangalagiu, V.; Amariucai-Mantu, D.; Antoci, V.; Giuroiu, C.L.; Mangalagiu, I.I. Hybrid Quinoline-Sulfonamide Complexes (M2+) Derivatives with Antimicrobial Activity. Molecules 2020, 25, 2946. [Google Scholar] [CrossRef]
  58. Al Matarneh, C.; Sardaru, M.; Apostu, M.; Rosca, I.; Ciobanu, C.; Mangalagiu, I.I.; Danac, R. Synthesis and antibacterial evaluation of new pyrrolo[3′,4′,3,4]pyrrolo[1,2a]quinoline derivatives. Studia UBB Chem. 2019, LXIV, 67–80. [Google Scholar] [CrossRef]
  59. Danac, R.; Al Matarneh, C.; Shova, S.; Daniloaia, T.; Balan, M.; Mangalagiu, I.I. New indolizines with phenanthroline skeleton: Synthesis, structure, antimycobacterial and anticancer properties. Bioorg. Med. Chem. 2015, 23, 2318–2327. [Google Scholar] [CrossRef]
  60. Danac, R.; Mangalagiu, I.I. Antituberculosis activity of nitrogen heterocycles derivatives: Bipyridine derivatives. Part III. Eur. J. Med. Chem. 2014, 74, 664–670. [Google Scholar] [CrossRef]
  61. Olaru, A.; Vasilache, V.; Danac, R.; Mangalagiu, I.I. Antimycobacterial activity of nitrogen heterocycles derivatives: 7-(pyridine-4-yl)- indolizine derivatives. Part VII. J. Enzym. Inhib. Med. Chem. 2017, 32, 1291–1298. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Hybrid drugs with various biological activities existing on the market.
Figure 1. Hybrid drugs with various biological activities existing on the market.
Pharmaceutics 14 02026 g001
Figure 2. Hybrid compounds with pyridine and quinoline skeleton used as antimicrobials existing in different clinical trial phases.
Figure 2. Hybrid compounds with pyridine and quinoline skeleton used as antimicrobials existing in different clinical trial phases.
Pharmaceutics 14 02026 g002
Scheme 1. Reaction pathway to obtain hybrid thiazole-pyridine 2ae and 4ae.
Scheme 1. Reaction pathway to obtain hybrid thiazole-pyridine 2ae and 4ae.
Pharmaceutics 14 02026 sch001
Scheme 2. Reaction pathways to obtain hybrid metal-pyridine derivatives 7.
Scheme 2. Reaction pathways to obtain hybrid metal-pyridine derivatives 7.
Pharmaceutics 14 02026 sch002
Scheme 3. Reaction pathway to obtain hybrid thiazolidine-pyridine 11af and 12ai.
Scheme 3. Reaction pathway to obtain hybrid thiazolidine-pyridine 11af and 12ai.
Pharmaceutics 14 02026 sch003
Scheme 4. Reaction pathway to obtain hybrid thiophen-pyrimidin-pyridine 1420.
Scheme 4. Reaction pathway to obtain hybrid thiophen-pyrimidin-pyridine 1420.
Pharmaceutics 14 02026 sch004
Scheme 5. Reaction pathway to obtain hybrid oxazino-pyridine 23aj.
Scheme 5. Reaction pathway to obtain hybrid oxazino-pyridine 23aj.
Pharmaceutics 14 02026 sch005
Scheme 6. Reaction pathway to obtain the tetrazolo-pyridine 25ad and tetrazolo-quinoline 26ae hybrids.
Scheme 6. Reaction pathway to obtain the tetrazolo-pyridine 25ad and tetrazolo-quinoline 26ae hybrids.
Pharmaceutics 14 02026 sch006
Scheme 7. Reaction pathway to obtain oxadiazolo-imidazopyridine hybrids 28aj.
Scheme 7. Reaction pathway to obtain oxadiazolo-imidazopyridine hybrids 28aj.
Pharmaceutics 14 02026 sch007
Scheme 8. Reaction pathway to obtain 1,3,4-triazolo-pyridine hybryds 30am and 31am.
Scheme 8. Reaction pathway to obtain 1,3,4-triazolo-pyridine hybryds 30am and 31am.
Pharmaceutics 14 02026 sch008
Scheme 9. Reaction pathway to obtain 1,2,3-triazolo-pyridine hybrids 35ar.
Scheme 9. Reaction pathway to obtain 1,2,3-triazolo-pyridine hybrids 35ar.
Pharmaceutics 14 02026 sch009
Scheme 10. Reaction pathway to obtain pyrazole-pyridine hybrids 3741.
Scheme 10. Reaction pathway to obtain pyrazole-pyridine hybrids 3741.
Pharmaceutics 14 02026 sch010
Scheme 11. Reaction pathway to obtain triazolo-pyridines 4245 and tetrazolo-pyridine 46 hybrids.
Scheme 11. Reaction pathway to obtain triazolo-pyridines 4245 and tetrazolo-pyridine 46 hybrids.
Pharmaceutics 14 02026 sch011
Scheme 12. Reaction pathway to obtain piperine-pyridine hybrids 48ah.
Scheme 12. Reaction pathway to obtain piperine-pyridine hybrids 48ah.
Pharmaceutics 14 02026 sch012
Scheme 13. Reaction pathway to obtain triazolo-quinoline hybrids 58aj and 59aj.
Scheme 13. Reaction pathway to obtain triazolo-quinoline hybrids 58aj and 59aj.
Pharmaceutics 14 02026 sch013
Scheme 14. Reaction pathway to obtain N-substituted-piperidino-quinoline hybrids 61a,b and1,2,3-triazolo-piperidino-quinoline hybrids 62ak.
Scheme 14. Reaction pathway to obtain N-substituted-piperidino-quinoline hybrids 61a,b and1,2,3-triazolo-piperidino-quinoline hybrids 62ak.
Pharmaceutics 14 02026 sch014
Scheme 15. Reaction pathway to obtain metal-quinoline hybrids 64ad.
Scheme 15. Reaction pathway to obtain metal-quinoline hybrids 64ad.
Pharmaceutics 14 02026 sch015
Scheme 16. Reaction pathway to obtain triazole-benzothiazole-quinoline hybrids 66af.
Scheme 16. Reaction pathway to obtain triazole-benzothiazole-quinoline hybrids 66af.
Pharmaceutics 14 02026 sch016
Scheme 17. Reaction pathway to obtaintriazole-quinoline hybrids 67au, 68az, 69an and 70a,b.
Scheme 17. Reaction pathway to obtaintriazole-quinoline hybrids 67au, 68az, 69an and 70a,b.
Pharmaceutics 14 02026 sch017
Scheme 18. Reaction pathway to obtain thiazole-quinoline hybrids 71 and 72.
Scheme 18. Reaction pathway to obtain thiazole-quinoline hybrids 71 and 72.
Pharmaceutics 14 02026 sch018
Scheme 19. Reaction pathway to obtain thiazolone-quinoline hybrids 7376.
Scheme 19. Reaction pathway to obtain thiazolone-quinoline hybrids 7376.
Pharmaceutics 14 02026 sch019
Scheme 20. Reaction pathway to obtain thiazole-quinoline hybrids 7782.
Scheme 20. Reaction pathway to obtain thiazole-quinoline hybrids 7782.
Pharmaceutics 14 02026 sch020
Scheme 21. Reaction pathway to obtain thiazole-quinoline hybrids 8385af.
Scheme 21. Reaction pathway to obtain thiazole-quinoline hybrids 8385af.
Pharmaceutics 14 02026 sch021
Scheme 22. Reaction pathway to obtain piperazin-quinoline hybrids 86al.
Scheme 22. Reaction pathway to obtain piperazin-quinoline hybrids 86al.
Pharmaceutics 14 02026 sch022
Scheme 23. Reaction pathway to obtain pyridine-quinoline hybrids 87aj.
Scheme 23. Reaction pathway to obtain pyridine-quinoline hybrids 87aj.
Pharmaceutics 14 02026 sch023
Scheme 24. Reaction pathway to obtain triazole-quinoline hybrids 88al.
Scheme 24. Reaction pathway to obtain triazole-quinoline hybrids 88al.
Pharmaceutics 14 02026 sch024
Scheme 25. Reaction pathway to obtain piperazin-quinoline hybrids 89aj.
Scheme 25. Reaction pathway to obtain piperazin-quinoline hybrids 89aj.
Pharmaceutics 14 02026 sch025
Scheme 26. Reaction pathway to obtainglycosylated-quinoline hybrids 9094.
Scheme 26. Reaction pathway to obtainglycosylated-quinoline hybrids 9094.
Pharmaceutics 14 02026 sch026
Scheme 27. Reaction pathway to obtain piperazine- and morpholine-quinoline hybrids 95ae and 96af.
Scheme 27. Reaction pathway to obtain piperazine- and morpholine-quinoline hybrids 95ae and 96af.
Pharmaceutics 14 02026 sch027
Scheme 28. Reaction pathway to obtain piperazino-quinoline hybrids 97100a,b.
Scheme 28. Reaction pathway to obtain piperazino-quinoline hybrids 97100a,b.
Pharmaceutics 14 02026 sch028
Scheme 29. Reaction pathway to obtain 7-substituted-quinoline hybrids 101104a,b.
Scheme 29. Reaction pathway to obtain 7-substituted-quinoline hybrids 101104a,b.
Pharmaceutics 14 02026 sch029
Scheme 30. Reaction pathway to obtain imidazolium-quinoline hybrids 105ah.
Scheme 30. Reaction pathway to obtain imidazolium-quinoline hybrids 105ah.
Pharmaceutics 14 02026 sch030
Scheme 31. Reaction pathway to obtain benzimidazole-quinoline and ferrocenyl-quinoline hybrids 106ae and 107ae.
Scheme 31. Reaction pathway to obtain benzimidazole-quinoline and ferrocenyl-quinoline hybrids 106ae and 107ae.
Pharmaceutics 14 02026 sch031
Scheme 32. Reaction pathway to obtain zwiterionic pyridine-fluoroquinolone and quinoline-fluoroquinolone hybrids 108ah and 109ah.
Scheme 32. Reaction pathway to obtain zwiterionic pyridine-fluoroquinolone and quinoline-fluoroquinolone hybrids 108ah and 109ah.
Pharmaceutics 14 02026 sch032
Scheme 33. Reaction pathway to obtain benzothiazole-benzo-quinoline hybrids 110, 111ad and 112am.
Scheme 33. Reaction pathway to obtain benzothiazole-benzo-quinoline hybrids 110, 111ad and 112am.
Pharmaceutics 14 02026 sch033
Scheme 34. Reaction pathway to obtain peptide-quinolone hybrids 113al.
Scheme 34. Reaction pathway to obtain peptide-quinolone hybrids 113al.
Pharmaceutics 14 02026 sch034
Scheme 35. Reaction pathway to obtain oxadiazole- and triazole-fluoroquinolone hybrids 114a,b and 115aj.
Scheme 35. Reaction pathway to obtain oxadiazole- and triazole-fluoroquinolone hybrids 114a,b and 115aj.
Pharmaceutics 14 02026 sch035
Scheme 36. Reaction pathway to obtain oxadiazole-fluoroquinolone hybrids 116at.
Scheme 36. Reaction pathway to obtain oxadiazole-fluoroquinolone hybrids 116at.
Pharmaceutics 14 02026 sch036
Scheme 37. Reaction pathway to obtain benzimidazole-quinoline hybrids 117ag, 118a,b and 119af.
Scheme 37. Reaction pathway to obtain benzimidazole-quinoline hybrids 117ag, 118a,b and 119af.
Pharmaceutics 14 02026 sch037
Scheme 38. Reaction pathway to obtain oxadiazole-quinoline hybrids 120ag.
Scheme 38. Reaction pathway to obtain oxadiazole-quinoline hybrids 120ag.
Pharmaceutics 14 02026 sch038
Scheme 39. Reaction pathway to obtain oxadiazole-quinoline hybrids 121ar.
Scheme 39. Reaction pathway to obtain oxadiazole-quinoline hybrids 121ar.
Pharmaceutics 14 02026 sch039
Scheme 40. Reaction pathway to obtain triazole-quinoline hybrids 122ac.
Scheme 40. Reaction pathway to obtain triazole-quinoline hybrids 122ac.
Pharmaceutics 14 02026 sch040
Scheme 41. Reaction pathway to obtain pyrazole-isoquinoline hybrids 123ag.
Scheme 41. Reaction pathway to obtain pyrazole-isoquinoline hybrids 123ag.
Pharmaceutics 14 02026 sch041
Scheme 42. Reaction pathway to obtain piperazine- and pyrimidine-isoquinoline hybrids 126ah and 127ah.
Scheme 42. Reaction pathway to obtain piperazine- and pyrimidine-isoquinoline hybrids 126ah and 127ah.
Pharmaceutics 14 02026 sch042
Scheme 43. Reaction pathway to obtain imidazole- and benzimidazole-quinoline hybrids 128134.
Scheme 43. Reaction pathway to obtain imidazole- and benzimidazole-quinoline hybrids 128134.
Pharmaceutics 14 02026 sch043
Scheme 44. Reaction pathway to obtain imidazole- and benzimidazole-pyridine hybrids 135138.
Scheme 44. Reaction pathway to obtain imidazole- and benzimidazole-pyridine hybrids 135138.
Pharmaceutics 14 02026 sch044
Scheme 45. Reaction pathway to obtain bis(imidazole)- and bis(benzimidazole)-pyridine hybrids 139143.
Scheme 45. Reaction pathway to obtain bis(imidazole)- and bis(benzimidazole)-pyridine hybrids 139143.
Pharmaceutics 14 02026 sch045
Scheme 46. Reaction pathway to obtain salts of imidazole- and benzimidazole-pyridine and quinoline hybrids 144147.
Scheme 46. Reaction pathway to obtain salts of imidazole- and benzimidazole-pyridine and quinoline hybrids 144147.
Pharmaceutics 14 02026 sch046
Scheme 47. Reaction pathway to obtain hybrid quinoline-sulfonamide complexes 149153.
Scheme 47. Reaction pathway to obtain hybrid quinoline-sulfonamide complexes 149153.
Pharmaceutics 14 02026 sch047
Scheme 48. Reaction pathway to obtain pyrrolo-quinoline and pyrrolo-isoquinoline hybrids 154ac and 155ac.
Scheme 48. Reaction pathway to obtain pyrrolo-quinoline and pyrrolo-isoquinoline hybrids 154ac and 155ac.
Pharmaceutics 14 02026 sch048
Scheme 49. Reaction pathway to obtain pyrrolo-phenanthroline hybrids 156ac.
Scheme 49. Reaction pathway to obtain pyrrolo-phenanthroline hybrids 156ac.
Pharmaceutics 14 02026 sch049
Scheme 50. Reaction pathway to obtain mono-indolizine-pyridine hybrids 157ae, salts of mono-indolizine -pyridine hybrids 159al and bis-indolizine-pyridine hybrids 160ad.
Scheme 50. Reaction pathway to obtain mono-indolizine-pyridine hybrids 157ae, salts of mono-indolizine -pyridine hybrids 159al and bis-indolizine-pyridine hybrids 160ad.
Pharmaceutics 14 02026 sch050
Table 1. Chemical structure, type of reactions and biological activity of the hybrids.
Table 1. Chemical structure, type of reactions and biological activity of the hybrids.
Synthesized HybridsType of ReactionsBiological Activity
Antimicrobial
Activity
Strains UsedEffect Observed
- thiazole-pyridine 2ae and 4ae- cyclocondensation- antibacterial;
- antifungal
- B. cereus, S. aureus, E. coli, P. aeruginosa;
- C. albicans
- active on B. cereus, S. aureus
- metal-pyridine derivatives 7- complexation- antibacterial;
- antifungal
- B. cereus, S. aureus, E. coli, P. aeruginosa;
- C. albicans
- active on C. albicans
- thiazolidine-pyridine 11af and 12ai- condensation- antitubercular- M. tuberculosis- active on Mtb
- thiophen-pyrimidin-pyridine 1420- cyclocondensation- antibacterial- S. aureus, S. mutans, E. coli,
K. pneumoniae
- active to all strains
-oxazino-pyridine 23aj- cyclocondensation, condensation- antibacterial;
- antifungal
- E. coli, P. aeruginosa, S. aureus,
S. pyogenes;
- C. albicans, A. niger, A. clavatus
- active on E. coli, C. albicans, A. clavatus
- tetrazolo-pyridine 25ad and
tetrazolo-quinoline 26ae
- cyclocondensation-antibacterial;
- antifungal
- K. pneumoniae, P. aeruginosa, S. aureus, S. pyogenes;
- C. albicans
- active on all bacterial strains
- oxadiazolo-imidazo-pyridine 28aj- cyclocondensation- antibacterial;
- antifungal
- E. coli, K. pneumoniae, S. aureus,
B. subtilis
- C. albicans, A. niger
- active on S. aureus,
C. albicans
- triazolo-pyridine 30an and 31an- cyclocondensation- antibacterial- S aureus, S. pyogenes, E. faecalis, E. coli, P. aeruginosa, A. baumannii- active on all strains
- triazolo-pyridine 35ar- cyclocondensation- antibacterial- E. coli, S. aureus, S. typhi- active on all strains
- pyrazole-pyridine 3741,
triazolo-pyridine 4245 and triazino-pyridine 46
- cyclocondensation- antibacterial;
- antifungal
- B. subtilis, E. coli, S. aureus, S. typhi
- C. albicans, A. niger
- active on all strains
- piperine-pyridine 48ah- acylation- antibacterial;
- antifungal
- B. subtilis, Streptobacillus, S. aureus,
E. coli, K. pneumoniae, P. aeruginosa,
E. faecalis, S. typhi
- A. niger, A. flavus, A. fumigatus, C. albicans
- active on E. coli,
K. pneumoniae, E. faecalis,
P. aeruginosa
- triazolo-quinoline 58aj and 59aj- reduction, N-alkylation, cyclocondensation- antibacterial;
- antifungal
- E. coli, P. aeruginosa, K. pneumoniae,
E. faecalis, S. aureus, S. pneumoniae,
B. subtilis
- C. albicans, C. parapsilosis
- active on all strains
- piperidino-quinoline 61a,b and triazolo-piperidino-quinoline 62ak- N-alkylation, cyclocondensation- antibacterial;
- antifungal
- E. coli, S. aureus;
- C. albicans
- active on all strains
- metal-quinoline 64ad- complexation- antibacterial;
- antifungal
- B. animalis, L. plantarum, B. subtilis,
S. aureus ATCC 663, S. aureus ATCC 25923, P. aeruginosa, P. mirabilis, E. coli, S. enterica;
- C. albicans, S. boulardii, A. flavus,
T. viridae, A. niger
- active on all strains
- triazole-benzothiazole-quinoline hybrids 66af- cyclocondensation- antibacterial;
- antifungal
- E. coli, B. subtilis, P. aeruginosa,
S. aureus;
- C. albicans, A. terreus
- active on all strains
- triazole-quinoline hybrids 67au, 68az,
69an and 70a,b
- cyclocondensation- antibacterial;
- antifungal
- E. coli, A. baumannii, K. pneumoniae,
S. aureus;
- C. albicans, C. neoformans
- active on all strains
- thiazole-quinoline 71, 72 and 7782,
thiazolone-quinoline 7376
- condensation, cyclocondensation- antibacterial;
- antifungal
- S. aureus, B. faecalis, B. subtilis, E. coli,
S typhi, P. aeruginosa;
- C. albicans, F. oxysporum
- active on S. aureus and E. coli
- piperazin-quinoline 86al,
- thiazole-quinoline 8385af
- alkylation, condensation- antibacterial;
- antifungal;
- antimalarial;
- antitubercular
- S. aureus, S. pyogenes, E. coli,
P. aeruginosa,
- C. albicans, A. niger, A. clavatus;
- P. falciparum;
- M. tuberculosis
- active on S. aureus and E. coli
- pyridine-quinoline 87aj- condensation- antibacterial;
- antifungal
- S. aureus, S. pyogenes, E. coli,
P. aeruginosa;
- C. albicans, A. niger, A. clavatus
- active on S. aureus,
E. coli, P. aeruginosa,
C. albicans
- triazole-quinoline 88al- cyclocondensation- antibacterial;
- antifungal
- S. aureus, S. pyogenes, E. coli,
P. aeruginosa;
- C. albicans, A. niger
- active on all strains
- piperazin-quinoline 89aj- cyclocondensation- antibacterial- S. aureus, MRSA, E. coli, P. aeruginosa- active on all strains
- glycosylated-quinoline hybrids 9094- hydrolysis, acylation- antibacterial;
- antifungal
- E. coli, L. monocytogenes, S. enterica,
P. aeruginosa, L. monocytogenes, MRSA, MSSA;
- C. albicans, A. flavus, F. solani,
S. chartarum, P. chrysogenum
- active on E. coli, C. albicans, P. chrysogenum
- piperazine- and morpholine-quinoline 95ae and 96af- condensation, alkylation- antibacterial;
- antitubercular
- A. baumanii, E. faecium, K. pneumonia, P. aeruginosa, E. coli, S. aureus
- M. tuberculosis
- active on S. aureus, E. coli,
A. baumanii and M. tuberculosis
- piperazino-quinoline 97104- acylation, alkylation- antibacterial- S. aureus, E. coli, P. aeruginosa,
B. subtilis
- active on all strains
- imidazolium-quinoline 105ah- substitution- antibacterial;
- antifungal;
- antitubercular
- S. aureus, E. coli, P. aeruginosa,
B. subtilis
- C. neoformans;
- M. tuberculosis
- active on C. neoformans
- benzimidazole-quinoline and ferrocenyl-quinoline 106ae and 107ae- cyclocondensation,
condensation
- antimalarial;
- antitubercular
- P. falciparum, P. berghei
- M. tuberculosis
- active on P. falciparum,
P. berghei
- zwiterionic pyridine-fluoroquinolone and quinoline-fluoroquinolone 108ah and 109ah- Mannich-electrophilic amination- antibacterial- S. aureus ATCC 6538, S. aureus MRSA N315, S. epidermidis ATCC 14990,
B. subtilis, E. coli, P. aeruginosa,
P. vulgaris, S. aureus MRSA 6347,
S. epidermidis MRSE, S. marcescens
- active on all strains
- benzothiazole-benzo-quinoline 110, 111ad and 112am.- cyclocondensation,
condensation
- antibacterial- S. aureus, E. coli, P. aeruginosa,
B. subtilis, E. faecalis, S. typhi
- active on S. aureus,
P. aeruginosa
- peptide-quinolone 113al
and conjugates with ciprofloxacine,
miofloxacine
- solid-phase peptide synthesis- antibacterial- A. baumanii, K. pneumonia,
P. aeruginosa, E. coli, S. aureus, MRSA, MSSA, MSSE, E. faecalis, E. cloacae,
S. maltophil
- only conjugates are active on all strains
- oxadiazole- and triazole-fluoroquinolone 114a,b and 115aj- one-pot three component Mannich reactions- antibacterial- S. aureus, E. coli, P. aeruginosa,
E. faecalis, K. pneumoniae, A. haemolyticus
- active on P. aeruginosa,
E. faecalis, A. haemolyticus
- oxadiazole-fluoroquinolone 116at- alkylation- antibacterial- S. aureus, MRSA- active on both
- benzimidazole-quinoline 117ag, 118a,b and 119af- alkylation- antibacterial;
- antifungal
- K. pneumonia, P. aeruginosa, E. coli,
S. aureus, S. aureus ATCC25923, S. aureus ATCC29213, E. faecalis, K. pneumonia,
P. aeruginosa ATCC27853, E. coli ATCC25922, A. baumanii, A. fumigatus, C. tropicalis, C. albicans, C. albicans ATCC90023, C. parapsilosis ATCC22019
- active on S. aureus,
K. pneumonia
- oxadiazole-quinoline 120ag- cyclocondensation- antibacterial;
- antifungal
- S. aureus, E. coli, B. cereus,
S. marcescens;
- A. niger, T. mentagrophytes, C. albicans, C. parapsilosis
- active on B. cereus
- oxadiazole-quinoline 121ar- acylation, cyclocondensation,
condensation
- leishmanicidal- Leishmania major- active
- triazole-quinoline 122ac- cyclocondensation- antifungal- C. albicans clinical strains and laboratory- active
- pyrazole-isoquinoline
123ag
- cyclocondensation- antibacterial;
- antifungal;
- antitubercular
- S. aureus, E. coli, E. faecalis, S. pyogens, V. cholera;
- C. albicans, C. tropicalis, C. parapsilosis, C. krusei, C. glabrata;
- M. tuberculosis
- active on S. aureus, V.
cholera, M. tuberculosis
- piperazine- and pyrimidine-isoquinoline 126ah and 127ah- N-alkylation, O-alkylation, S-alkylation- antibacterial;
- antifungal;
- antitubercular
- S. aureus, E. coli, K. pneumonia,
B. subtilis;
- C. albicans, A. niger, A. oryzae,
P. chrysogenum;
- M. tuberculosis
- active on all strains
- imidazole- and benzimidazole-quinoline 128134- cycloaddition, N-alkylation- antibacterial;
- antifungal
- S. aureus, E. coli;
- C. albicans
- active on all strains
- imidazole- and benzimidazole-pyridine 135138- cycloaddition, N-alkylation- antibacterial;
- antifungal
- S. aureus, E. coli;
- C. albicans
- active on S. aureus, E. coli
- bis(imidazole)- and bis(benzimidazole)-pyridine 139143- N-alkylation- antitubercular- M. tuberculosis- active
- imidazole- and benzimidazole-pyridine and quinoline 144147- N-alkylation- antitubercular- M. tuberculosis- no activity
- quinoline-sulfonamide complexes 149153- acylation, complexation- antibacterial;
- antifungal
- S. aureus, E. coli;
- C. albicans
- active on all strains
- pyrrolo-quinoline and pyrrolo-isoquinoline 154ac and 155ac- cycloaddition- antibacterial;
- antifungal
- S. aureus, E. coli;
- C. albicans
- no activity
- pyrrolo-phenanthroline 156ac- cycloaddition- antitubercular- M. tuberculosis- active
- mono-indolizine-pyridine 157ae, salts of mono-indolizine-pyridine 159al and bis-indolizine-pyridine 160ad- N-alkylation, cycloaddition- antitubercular- M. tuberculosis- active
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Amariucai-Mantu, D.; Mangalagiu, V.; Bejan, I.; Aricu, A.; Mangalagiu, I.I. Hybrid Azine Derivatives: A Useful Approach for Antimicrobial Therapy. Pharmaceutics 2022, 14, 2026. https://doi.org/10.3390/pharmaceutics14102026

AMA Style

Amariucai-Mantu D, Mangalagiu V, Bejan I, Aricu A, Mangalagiu II. Hybrid Azine Derivatives: A Useful Approach for Antimicrobial Therapy. Pharmaceutics. 2022; 14(10):2026. https://doi.org/10.3390/pharmaceutics14102026

Chicago/Turabian Style

Amariucai-Mantu, Dorina, Violeta Mangalagiu, Iustinian Bejan, Aculina Aricu, and Ionel I. Mangalagiu. 2022. "Hybrid Azine Derivatives: A Useful Approach for Antimicrobial Therapy" Pharmaceutics 14, no. 10: 2026. https://doi.org/10.3390/pharmaceutics14102026

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop